You are on page 1of 47

 .

BOBHFNFOUPG)FQBUJUJT$BOE
 )*7$PJOGFDUJPO
 $MJOJDBM1SPUPDPMGPSUIF8)0&VSPQFBO3FHJPO

Contents

I. Epidemiology and natural history of HCV in HIV infection .............................................. 229


1. Prevalence, risk factors and transmission ................................................................................. 229
1.1. Prevalence of HCV in HIV infection ............................................................................. 299
1.2. Primary modes of transmission ...................................................................................... 231
1.3. Genotypes ...................................................................................................................... 231
2. Access of coinfected patients to hepatitis C treatment .......................................................... 232
3. Reciprocal influences of HIV and HCV ................................................................................ 232
3.1. Impact of HIV infection on HCV disease progression .................................................. 232
3.2. Impact of HCV infection on HIV disease progression .................................................. 233

II. Identification of HCV/HIV.................................................................................................... 234


1. Assessment of HCV risk and diagnosis of hepatitis C in HIV-infected patients ................. 234
1.1. Initial laboratory assessment of HCV status ................................................................. 234
1.2. Evaluation of HCV disease severity .............................................................................. 235
1.2.1. Clinical evaluation of liver disease ...................................................................... 235
1.2.2. Biochemical parameters ....................................................................................... 235
1.2.3. Child-Pugh score .................................................................................................. 236
1.2.4. Ultrasound ............................................................................................................ 236
1.2.5. Histological evaluation ........................................................................................ 236
1.2.6. Non-invasive markers of liver fibrosis ................................................................. 237
1.2.7. Clinical situations not requiring histological evaluation ..................................... 237
1.3. Evaluation of comorbidities and co-conditions ............................................................. 238
1.3.1. Psychiatric disorders ............................................................................................ 238
1.3.2. Alcohol abuse ....................................................................................................... 238
1.3.3. Drug use ............................................................................................................... 238
1.3.4. Other comorbidities and co-conditions ................................................................ 238
1.4. Evaluation and treatment algorithms for hepatitis C ..................................................... 240
1.4.1. Algorithm 1 .......................................................................................................... 240
1.4.2. Algorithm 2 .......................................................................................................... 242
2. Assessment of HIV risk and diagnosis of HIV/AIDS in HCV patients ............................... 243

III. Clinical management of HCV/HIV patients ...................................................................... 244


1. Coinfected patients not requiring any treatment ................................................................ 244
2. Coinfected patients requiring only HCV treatment ............................................................ 244
2.1. Indications for HCV treatment .................................................................................... 244
2.2. Predictors of sustained virological response probability ............................................ 244
2.3. Contraindications for hepatitis C treatment ................................................................ 245
2.4. Treatment of acute hepatitis C ..................................................................................... 245
2.5. Treatment of chronic hepatitis C (doses and schedules) ............................................. 245
2.6. Treatment duration ...................................................................................................... 246
3. Coinfected patients requiring only HIV/AIDS treatment................................................... 246
3.1. Initiation of HAART ................................................................................................... 246
3.2. Considerations in choosing a HAART regimen ........................................................... 247
3.3. First-line HAART regimens ......................................................................................... 247
3.4. Second-line HAART regimens .................................................................................... 248
4. Coinfected patients requiring both HCV and HIV/AIDS treatment ................................... 248
4.1. Strategy for initiation of treatment ............................................................................... 248
4.2. Considerations of ARVs when treating both HCV and HIV infections ....................... 249
4.3. Hepatotoxicity of ARV drugs ....................................................................................... 250
4.4. ARV dose adjustment in patients with cirrhosis .......................................................... 250
5. Clinical monitoring ............................................................................................................. 252
5.1. Virological response monitoring .................................................................................. 252
5.2. Histological response monitoring ................................................................................ 252
5.3. Tolerance monitoring ................................................................................................... 253
5.4. Management of toxicity and side-effects of PEG-IFN + RBV treatment .................... 253
5.4.1. Anaemia and neutropenia ................................................................................... 253
5.4.2. Dose adjustment of PEG-IFN and RBV ............................................................. 253
5.4.3. Influenza-like symptoms..................................................................................... 254
5.4.4. Nausea ................................................................................................................ 254
5.4.5. Depression .......................................................................................................... 254
5.4.6. Dysthyroidism .................................................................................................... 254
5.5. Management of treatment adherence ........................................................................... 254
5.6. Management of non-responders ................................................................................... 255
5.7. Management of end-stage liver disease ....................................................................... 255
5.7.1. Testing for hepatocellular carcinoma .................................................................. 255
5.7.2. Testing for oesophageal varices .......................................................................... 255
5.8. Drug–drug interactions ................................................................................................ 256
5.8.1. Interactions between HIV drugs and HCV drugs ............................................... 256
5.8.2. Interactions among recreational drugs, OST, anti-HCV drugs and ARVs .......... 256
5.9. Hepatotoxicity of TB drugs in chronic HCV infection ................................................ 256

IV. Suggested minimum data to be collected at the clinical level ........................................... 257

Annex 1. Laboratory assays for HCV ....................................................................................... 258

Annex 2. Alternative biochemical tests to assess hepatic fibrosis ........................................... 260

Annex 3. Alcohol screening questionnaires............................................................................... 261

Annex 4. Management of end-stage liver disease ..................................................................... 263

Annex 5. Research needs and alternative treatments .............................................................. 265

References .................................................................................................................................... 267


Management of Hepatitis C and HIV Coinfection

I. Epidemiology and natural history of HCV in HIV infection

In Europe, the prevalence of hepatitis C virus (HCV) infection in HIV-infected patients is particu-
larly high – and still rising, in contrast to the rest of the world. Yet only a minority of HCV/HIV-
coinfected patients are treated for their hepatitis. The compounding effect of coinfection makes the
care for these patients a major challenge.

In the pre-HAART era, the late consequences of HCV-related chronic liver disease in coinfected
individuals were overshadowed by AIDS mortality connected with severe immune deficiency. With
the development of HAART, morbidity and mortality among HIV-infected patients have decreased
significantly. The consequences of liver-related disease associated with chronic HCV infection are
now far more worrying. End-stage liver disease (ESLD) is now the predominant cause of death in
patients coinfected by HCV and HIV, as well as in hepatitis B virus (HBV)/HIV-coinfected patients
(1), despite the availability of treatments with proven efficacy (2–5). Most patients are, however,
not treated, underscoring the need for treatment guidelines. Efforts must also be made, via multidis-
ciplinary health-care services, to increase the applicability and availability of treatment, especially
in more vulnerable populations, including but not limited to migrants, injecting drug users (IDUs),
prisoners, people with psychiatric illnesses and people who consume too much alcohol.

1. Prevalence, risk factors and transmission


Worldwide about 180 million people are chronic carriers of HCV. Overlapping routes of transmis-
sion for HCV and HIV result in a high frequency of coinfection in Europe.

1.1. Prevalence of HCV in HIV infection


The prevalence of HCV infection in individuals infected with HIV in the WHO European Region
is very high, averaging 40% and reaching 50–90% in urban areas. Data from a EuroSIDA study
(see Fig. 1) shows the prevalence is higher in the eastern (47.7%) and southern (44.9%) EuroSIDA
regions than in the northern (24.5%) EuroSIDA region, due to the high rates of injecting drug use
in the two former regions (6).

Fig. 1. Prevalence of HCV in HIV-infected patients in Europe

North: 24.5 %

East: 47.7 %
Central: 22.9 %

South: 44.9 %

Source: Rockstroh et al. (7).

229
HIV/AIDS TREATMENT AND CARE CLINICAL PROTOCOLS FOR THE WHO EUROPEAN REGION

The prevalence of HCV antibodies also varies widely among HIV transmission groups, ranging
from 7–8% in men who have sex with men to 60–70% in haemophiliacs and 80–90% in IDUs, the
most important group (see Fig. 2) (8–12). HCV is easily transmitted among IDUs, which makes it
difficult to prevent. IDU transmission occurs in several ways:
• sharing needles and syringes
• sharing auxiliary paraphernalia, such as cookers, straws, swabs, tourniquets and cotton
• sharing drug doses from a common syringe
• accidental needle-sticks.

Fig. 2. Prevalence of HCV antibodies in different transmission groups

HCV
HBS ag
% Positive

Source: Alter (13).

The prevalence of HCV among IDUs increases with the duration of injection, as shown in Fig. 3.

Fig. 3. Prevalence of HCV among IDUs in relation to injecting history

HCV
HBS ag

Source: Quaglio et al. (14).

230
Management of Hepatitis C and HIV Coinfection

1.2. Primary modes of transmission


The primary modes of transmission for HCV are parenteral and vertical (from mother to child); it is
rarely transmitted sexually. In Europe, the most common route of transmission occurs via injecting
drug use. Although sexual transmission of HCV occurs in <1% (15) of monogamous couples, there
have been increasing reports of sexual transmission between men who have sex with men (MSM)
HCV
HBS ag Household contact with an HCV-infected person has been associated with an average non-
(16).
sexual transmission rate of 4% (0–11%) (17). Other risk factors for transmission of HCV include
tattooing and accidental needle-sticks in medical settings (18).

1.3. Genotypes
HCV exhibits a high genetic heterogeneity around the world, with six different clades or genotypes
being distinguished and differing as much as 30% in their genome (see Fig. 4). Furthermore, phylo-
genetic analyses can also distinguish subtypes and isolates within a particular type.

Fig. 4. Phylogenetic tree of HCV genotypes and subtypes

Source: Francisus (19).

HCV
From an epidemiological point of view, infection with genotypes 3 and 4 is more prevalent in IDUs
and HIV-coinfected patients than in monoinfected patients. Acute
HBS aggenotype 4 infection has recently
been found among MSM (16).
% Positive

The distribution of genotypes may differ from one region of the world to another. As genotypes
have differed in their sensitivity to the standard treatment since 2005 – pegylated interferon (PEG-
IFN) and ribavirin (RBV) – it is important to know the genotype of each patient and the distribution
of the genotypes in each country.

Fig. 5. Prevalence of genotypes 1–3 in the United States and western Europe

Source: Simmonds et al., Zeuzem S et al. (20, 21).

231
HIV/AIDS TREATMENT AND CARE CLINICAL PROTOCOLS FOR THE WHO EUROPEAN REGION

2. Access of coinfected patients to hepatitis C treatment


Low percentages (0–23%) of coinfected patients have access to hepatitis C treatment (22). There
may be several reasons for this:
• The efficacy of PEG-IFN and RBV in treating coinfected patients was only published in 2004,
and these drugs are not widely available.
• A great number of patients who continue active drug use do not have access to substitution treat-
ment and/or ART.
• Many countries lack guidelines for diagnosis and treatment.
• Evaluation of the severity of HCV disease and treatment requires high technology and skills.
• Neuropsychological side-effects and toxicity are frequent during HCV treatment.
• Treatment is very costly.

3. Reciprocal influences of HIV and HCV

3.1. Impact of HIV infection on HCV disease progression


• Several studies have demonstrated that patients coinfected with HCV and HIV have more rapid
fibrosis progression than monoinfected patients, even after taking into account age, sex and al-
cohol consumption (23).
• People with HCV/HIV coinfection may have quantitative and/or qualitative deficiency in their
immune responses to HCV. HIV accelerates the course of HCV-associated liver disease, particu-
larly in patients who are more severely immune deficient, by increasing:
° the HCV viraemia level from two- to eightfold, resulting in a significant decrease in sponta-
neous recovery from acute hepatitis (24);
° the risk of mother-to-child and sexual transmission (from averages of 6% to 20% and from
0% to 3%, respectively); and
° rates of liver fibrosis (two- to fivefold), cirrhosis, decompensation, hepatocellular carcinoma
(HCC) and liver-related mortality (25).
• Liver disease is the leading cause of morbidity and mortality in HCV/HIV-coinfected patients in
some parts of Europe, despite the suggestion that HAART, especially protease inhibitors, may
decrease the severity of liver disease and the related mortality (1).
• Comorbidities with hepatic consequences (drug hepatotoxicity, HBV, steatosis, alcohol or drug
abuse) are frequent in coinfected patients and may increase the rate of complications associated
with HCV-related liver disease. Patients with CD4 <200 cells/mm3 are those most likely to prog-
ress to severe liver disease (6, 23, 25, 26). For example, HIV-infected patients with CD4 <200
cells/mm3 who drink more than 50 g of alcohol daily have a median expected time to cirrhosis
of 16 years, versus 36 years for HIV-infected patients with CD4 >200 cells/mm3 who drink 50 g
or less of alcohol daily (26).
• Spontaneous clearance of HCV is significantly lower in HIV-infected patients than in immuno-
competent patients with acute hepatitis. As HCV ribonucleic acid (RNA) might become tempo-
rarily undetectable during the acute phase of HCV infection, clearance must be confirmed with
a sensitive qualitative HCV RNA assay on at least two occasions six months apart (27, 28).
• In profoundly immunosuppressed patients, HCV serology has occasionally been found to be
falsely negative despite HCV chronic infection. Such false negatives have become very rare due
to the high sensitivity of third-generation serology (27, 28).

1
HCV RNA testing should, therefore, be performed in people at risk, such as IDUs and MSM, and in others who may be
profoundly immunosuppressed and present unexplained ALT elevation despite negative HCV serology.

232
Management of Hepatitis C and HIV Coinfection

3.2. Impact of HCV infection on HIV disease progression


HCV has little or no effect on the response to ART or on immunological, virological or HIV-related
clinical disease progression. Although HCV antibodies per se do not influence progression, infec-
tion with certain multiple genotypes might do so (29).

Extended follow-up in various studies indicate that patients on HAART do not have any major dif-
ferences in HIV-related mortality from HCV/HIV-coinfected patients or those infected with HIV
alone, particularly if ART is given (6). There is, however, an increased risk for liver disease-related
morbidity and mortality in hepatitis-coinfected HIV, as well as more hepatotoxicity under ART
regimens (30).

233
HIV/AIDS TREATMENT AND CARE CLINICAL PROTOCOLS FOR THE WHO EUROPEAN REGION

II. Identification of HCV/HIV


1. Assessment of HCV risk and diagnosis of hepatitis C in HIV-infected
patients

1.1. Initial laboratory assessment of HCV status

1.1.1. Step 1: All HIV-infected patients should be tested for HCV antibodies.
• For patients with acute HCV infection, it is important to bear in mind that antibodies may not
be detectable for three to eight weeks following initial HCV infection. Retesting is not neces-
sary if the infection was transmitted heterosexually and in the absence of other risky behaviour.
For others who continue to run the risk of infection, such as active IDUs or MSM with multiple
partners, testing is recommended every one to two years (31).
• The presence of HCV antibodies is indicative of past or present infection. Antibodies persist
indefinitely, in chronically infected patients but the antibody titres may decrease (and even dis-
appear) in patients who clear HCV (either spontaneously or after antiviral treatment).
• HIV infection can impair antibody responses to HCV infection (27), so a second- or third- gen-
eration enzyme immunoassay (EIA) for HCV antibodies should be used in coinfected individuals.
• In HCV antibody-negative HIV patients with profound immunosuppression, HCV RNA deter-
mination is recommended when there are liver test abnormalities or clinical suspicion of liver
disease.

1.1.2. Step 2: When testing for HCV antibodies is positive, detection of HCV RNA should be performed
to confirm or exclude active replication.
• HCV RNA can be detected as soon as a few days after infection.
• HCV RNA can be detected by PCR (polymerase chain reaction) or by TMA (transcription-me-
diated amplification).
• Persistence of HCV RNA more than six months after initial infection confirms chronic hepatitis
C (27, 31).
• Determination of HCV RNA can be done through qualitative or quantitative assays.
° A qualitative assay is enough for diagnostic purposes.
° A quantitative assay (viral load) is important for assessment of patients who will receive
HCV treatment.
• High pretreatment HCV RNA levels are associated with lower rates of sustained virological
response (SVR); the cut-off is generally 800 000 copies/ml (IU/ml) (32). SVR rates may reach
60% in persons with either a genotype other than 1 or 4, or genotype 1 HCV infection with an
HCV RNA level ≤800 000 IU/ml after 48 weeks of PEG-IFN and RBV treatment, as opposed
to only 18% for those with genotype 1 and an HCV RNA level >800 000 IU/ml. (2–5, 32).
• It is important to consider that viral load is higher (0.5–1 log on average) in HCV/HIV-coin-
fected individuals than in those who are monoinfected. This may also account for higher HCV
transmission to children born to coinfected mothers. Therefore, assays with a wide dynamic
range may represent an advantage.

1.1.3. Step 3: Use HCV genotype determination in predicting treatment response.


Distribution of genotypes differs between HCV-monoinfected and coinfected patients, as illustrated
in Table 1.

234
Management of Hepatitis C and HIV Coinfection

Table 1. Distribution of genotypes by monoinfection and coinfection, in %

Genotype 1 Genotype 2 Genotype 3 Genotype 4


Monoinfected 65 12 19 3
Coinfected 60 5 28 8

Source: Fried et al., Tottiani et al. (33, 34).

• Infections with more than one HCV genotype appear to be more often (>5%) in patients coin-
fected with HCV and HIV, particularly IDUs and haemophiliacs (29, 35).
• HCV genotype plays a predominant role as a predictor of SVR in HIV-infected patients, as it has
been found in all studies of people without HIV infection.
° For genotypes other than 1 or 4, SVR rates are generally high, ranging from 73% in the
ACTG 5071 study (4) to 62% in the APRICOT study (3), 53% in the Barcelona study (5) and
44% in the RIBAVIC study (2).
° For genotype 1, SVR rates range from 29% in APRICOT (3) to 17% in RIBAVIC (2) and
14% in ACTG 507 (4), while Barcelona reported a 38% SVR rate for those with genotype 1
or 4 (5).

For more information about laboratory assays for HCV, please see Annex 1.

1.2. Evaluation of HCV disease severity


• Evaluation of HCV disease severity should include attempting to define the duration of the in-
fection. The date of infection is usually defined as the first date of risk exposure to HCV infec-
tion (first drug injection date, etc.).
• For decisions regarding treatment, the focus of the evaluation should be on chronic liver disease,
comorbidities and co-conditions.

1.2.1. Clinical evaluation of liver disease


Clinical signs of cirrhosis are:
• stellar angiomas
• dysmorphic liver
• digital hippocratism (clubbing of the fingers)
• collateral abdominal circulation
• signs of hepatic decompensation (ascites, icterus, encephalopathy, etc.).

1.2.2. Biochemical parameters


Biochemical tests to be performed are:
• transaminases (ALT, AST), 
• gamma glutamyl transpeptidase (GGT) (may increase in case of cirrhosis)
• alcalines phosphatases (to establish another possible cause of hepatic disease)
• bilirubine
• albumin
• prothrombin time.

2
Alanine aminotransferase (ALT) levels do not necessarily reflect the stage of fibrosis, especially in HCV/HIV-coinfected pa-
tients. A normal ALT level alone should not be grounds to defer treatment. A biopsy in this situation can help to make a more
informed decision. In the RIBAVIC study, baseline ALT >3 times the upper limit of normal was a predictor of higher SVR.
3
Asparate aminotransferase (AST) levels should be controlled when performing the initial complete hepatic evaluation to elimi-
nate other causes of hepatic disease; for example, in cases of alcoholic intoxication there may be an increase in AST and GGT.

235
HIV/AIDS TREATMENT AND CARE CLINICAL PROTOCOLS FOR THE WHO EUROPEAN REGION

1.2.3. Child-Pugzh score


The Child-Pugh Score, combining clinical symptoms and biological tests (Table 2), is useful for
grading the severity of ESLD and should be performed in all patients with cirrhosis (36).

Table 2. Child-Pugh classification

Points
Clinical and biochemical parameters
1 2 3
<2 mg/dl 2–3 mg/dl >3 mg/dl
(<34 µmol/l) (34–50 µmol/l) (>50 µmol/l)
Bilirubin

Albumin >3.5 g/dl 2.8–3.5 g/dl <2.8 g/dl


Ascites Absent Moderatea Severe/ refractoryb
Encephalopathy Absent Moderate (stage I–II) Severe (stage III–IV)
Prothrombin time c
>60% 40–60% <40%
a
Controlled medically.
b
poorly controlled.
c
now replaced in some European countries by international normalized ratio (INR with the following Child-Pugh values:
INR <1.70 = 1 point; 1.71–2.20 = 2 points; >2.20 = 3 points.
Source: Pugh et al. (36).

Interpretation of the Child-Pugh classification:


• Class A (5–6 points) – compensated cirrhosis
• Class B (7–9 points) – compensated cirrhosis
• Class C (10–15 points) - decompensated cirrhosis

1.2.4. Ultrasound
Ultrasound (Doppler if possible) examination of the liver can reveal:
• cirrhosis: dysmorphy of the liver
• steatosis: hyperechogenic liver
• possibly early HCC: nodular unique or, rarely, multiple lesions.

1.2.5. Histological evaluation


Liver biopsy is the standard procedure for evaluation of the severity of liver disease (see Table 3 for
indications). It is especially important for patients with a suspected low chance of SVR (genotype 1
with a high viral load) or excess risk of severe side-effects, and allows evaluating:
• the degree of fibrosis and necroinflammatory activity
• the presence of comorbidities (steatosis, drug toxicity, alcohol related lesions, HBV).

Table 3. Indications for liver biopsy in HCV/HIV-coinfected patients

Indications for biopsy Biopsy not required


Genotype 1 or 4 with high HCV viral load Genotype 2 and 3
(>800 000 IU/ml)
Genotype 1 (and probably 4) with low HCV load
Presence of comorbidities: (≤800 000 IU/ml)
- excessive alcohol consumption
- coinfection with HBV and/or hepatitis delta virus Clinical signs of cirrhosis
- suspicion of medication-associated hepatotoxicity

Biopsies must be performed by trained physicians, as significant complications may occur in 1/200
patients. They should be read by specialized anatomopathologists, as subtle differences may change

236
Management of Hepatitis C and HIV Coinfection

the classification of the severity of the disease. These limitations impede generalized biopsies for all
HCV-infected patients (see section II.1.2.7 below for clinical situations not requiring liver biopsy).
Activity and fibrosis are two major histological features of chronic hepatitis C that are included in
proposed classifications, such as Ishak, Metavir and Knodell, that allow improved consistency in
interpretation of hepatic fibrosis with a somewhat weaker reproducibility for hepatic inflammation
grade (37, 38). See Table 4.

Table 4. Metavir classification: activity and fibrosis scoring

Lobular necrosis
Activity score (A)
Absent (0) Moderate (1) Severe (2)
Absent (0) A0 A1 A2
Parcellar Minimal (1) A1 A1 A2
necrosis Moderate (2) A2 A2 A3
Severe (3) A3 A3 A3

A0 = no histological activity; A1 = minimal activity; A2 = moderate activity; A3 = severe activity.

Table 4a.

Fibrosis score (F)


F0: absence of portal fibrosis
F1: stellar portal fibrosis with no septa
F2: portal fibrosis with some septa
F3: many septa but no cirrhosis
F4: cirrhosis

Source: Simmonds et al. (20).

This system assesses histological lesions in chronic hepatitis C using two separate scores, one for
necroinflammatory grade (A for Activity) and another for the stage of fibrosis (F). The fibrosis
stage and inflammatory grade are correlated, but for approximately one third of patients there is
discordance. In lower grades of liver fibrosis (F0–F1), regardless of HCV genotype, treatment can
be deferred. See Table 4a.

1.2.6. Non-invasive markers of liver fibrosis


Non-invasive tools for assessing liver fibrosis, such as those based on serum markers (for example,
FibroTest™) or image technique (for example, FibroScan™) are available. Several non-invasive
methods to evaluate inflammation and fibrosis have been developed for monoinfected patients and
include serological tests combining serum fibrosis markers. They are used to distinguish Metavir
fibrosis stages 0–2 from stages 3 and 4. The tests are quite reliable, are better accepted by patients
than biopsies and could potentially save approximately 50% of patients from being biopsied.

Recently, alternatives to biopsies have become available for coinfected patients (39), including a
combination of biochemical tests indicating the degree of liver inflammation and fibrosis, such as
the Forns index which has been recently validated for HIV/HCV-coinfected patients (40), and an
elastometric method reflecting the degree of fibrosis (see Annex 2) (41, 42).

237
HIV/AIDS TREATMENT AND CARE CLINICAL PROTOCOLS FOR THE WHO EUROPEAN REGION

1.2.7. Clinical situations not requiring histological evaluation


The First European Consensus Conference on the Treatment of Hepatitis in HIV-Infected Patients
did not mandate biopsy in cases where treatment is already indicated (43). Treatment without bi-
opsy or other liver assessment is recommended in the following situations:
• infection with HCV genotype 2 or 3;
• infection with HCV genotype 1 with a low viral load; and
• absence of major contraindications and patient willingness to undergo treatment, in which case
the SVR will be on the order of 40–60% (2–5).

Given the limitations of biopsy and the faster progression of fibrosis in HCV/HIV patients, treat-
ment should still be offered when candidates for biopsy decline it or lack access to it.

1.3. Evaluation of comorbidities and co-conditions

1.3.1. Psychiatric disorders


• An initial evaluation of psychiatric disorders should be performed, as treatment with IFN can
reveal and worsen depression. Treatment for hepatitis C should therefore be deferred in patients
with moderate to severe depression until the condition improves. Prophylactic treatment with
psychiatric drugs may be advisable and treatment may be feasible thereafter.
• In patients with mild psychiatric illness, treatment for hepatitis C should not be deferred and
counselling and/or antidepressant medication should be offered along with HCV treatment.

1.3.2. Alcohol abuse


• Assessment of alcohol intake is an important part of evaluation (please see Annex 3).
• Heavy alcohol intake (50 g/day or more) contributes to fibrosis of the liver, which can be identi-
fied by biopsy in HCV patients independently of other predictors. This intake is equivalent to
five or more drinks per day, in which a drink = 10 g of alcohol, for example 330 ml (12 oz) of
beer, 150 ml (5 oz) of wine or 38 ml (1.25 oz) of hard alcohol.
• There is evidence of synergistic interaction between alcohol consumption ≥80 ml/day and
chronic HBV or HCV infection (44). Continued alcohol consumption increases HCV replica-
tion, accelerates fibrogenesis and liver disease progression in hepatitis B and C and diminishes
the response and adherence to treatment (especially if consumption is >50 g/day).
• Active alcohol intake is considered a relative contraindication for IFN-based treatment, due
to the documented non-compliance of heavy drinkers in medical therapies, combined with the
side-effects that otherwise affect compliance (45).
• Psychological, social and medical support should be offered to reduce alcohol intake to <10
g/day or stop it altogether.

1.3.3. Drug use


• Treatment of patients on opioid substitution therapy should not be deferred.
• Initiation of HCV treatment in active drug users should be considered on a case-by-case basis.
(Please refer to Protocol 5, HIV/AIDS treatment and care for injecting drug users.)
• Medical, psychological and social support from a multidisciplinary team should be provided for
these patients.

238
Management of Hepatitis C and HIV Coinfection

1.3.4. Other comorbidities and co-conditions


Testing of comorbidities should include a comprehensive history with a particular focus on factors
associated with more progressive liver injury. Analysis can include:
• testing for viral liver diseases
• testing for tuberculosis (TB) and sexually transmitted infections (STIs) that need treatment be-
fore HCV treatment begins.

When a treatment has been decided, other tests are needed:


• thyroid-stimulating hormone (TSH) dosage;
• dosage of antiperoxydase, antinuclear, anti-smooth muscle, anti-liver-kidney microsome anti-
body (LKM1);
• creatininaemia;
• proteinuria ;
• glycaemia;
• ferritinaemia;
• electrocardiogram (ECG, to detect coronary disease that could decompensate after treatment-
induced anaemia);
• a pregnancy test.

4
  For HBV and HAV please refer to Protocol 7, Management of hepatitis B and HIV coinfection.
5
  See Protocol 4, Management of tuberculosis and HIV coinfection, and the European STD Guidelines (46).
6
  It should be explained that because RBV is teratogenic and contraindicated during pregnancy, procreation should be avoid-
ed during treatment and six months after, and that due to higher levels of HCV viraemia in coinfected women, approximately
20% transmit HCV to their offspring, versus 7–8% in those monoinfected with hepatitis C (47).

239
HIV/AIDS TREATMENT AND CARE CLINICAL PROTOCOLS FOR THE WHO EUROPEAN REGION

1.4. Evaluation and treatment algorithms for hepatitis C

1.4.1. Algorithm 1
This algorithm is preferred and focuses on genotyping.

Fig. 6. Algorithm 1

HIV- and HCV-positive


serology

HCV RNA-positive Child-Pugh B or C


(qualitative or quantitative) decompensated cirrhosis

Genotyping

1 or 4 2 or 3

HCV RNA HCV RNA


(quantitative) (quantitative)

High Low
>800 000 ≤800 000

Liver biopsy or
Not available
non-invasive markersa

Moderate or Treatment during 12


Mild
severe weeks

Observe, re-evaluate HCV RNA


after three years (quantitative)

Continue Consider liver


treatment >2 log drop Stop transplantation where
<2 log drop
until 48 or negative treatment available, otherwise
weeks refer to palliative care

a
FibroScan (image technique), Fibro Test (serum fibromarkers)

240
Management of Hepatitis C and HIV Coinfection

In Algorithm 1, the decision to treat lies mainly upon the HCV genotype determination and HCV
quantification. Liver biopsy is limited to patients with genotype 1, high viral load and low response
to PEG-IFN and RBV.
• Subsequent to an HCV/HIV positive serology, qualitative HCV RNA detection should be under-
taken to confirm the chronicity of hepatitis.
• In case of positive HCV RNA, a genotyping should be performed.
• In case of genotype 2 or 3, more frequently found in IDUs, treatment should be proposed for all
patients without liver biopsy where there is no contraindication (please see contraindications in
section III.2.3).
• In case of genotype 1, the patient should have a quantification of HCV RNA, since responses
are related to viral load. This test should be available everywhere HIV viral load is performed.
• In the absence of local testing possibilities, the patient should be referred to a specialist, or a
sample should be collected at the district level and a genotyping test done centrally.
° When viral load is low (≤800 000 IU/ml), treatment of genotype 1 is recommended without
a liver biopsy.
° When viral load is high (>800 000 IU/ml), an assessment of liver fibrosis by biopsy is rec-
ommended to differentiate patients with severe liver disease.
• A fibrosis score of F2­–F4 indicates a need for immediate treatment.
• Mild liver disease (F0, F1) indicates that treatment should be delayed due to the low chances of
SVR.
• Follow-up treatment should rely on HCV RNA quantification at week 12, and then HCV RNA
qualitative detection at weeks 24 and 48.
° At week 12, if the drop of viral load is less than 2 log, the treatment should be stopped be-
cause the chance of success does not exceed 1–2% regardless of genotype. Otherwise, the
treatment should be continued.
° Additional qualitative tests should be performed at week 24 and treatment should be stopped
if HCV viral load is detectable; otherwise, treatment should be pursued until week 48 and
treatment efficacy checked with a qualitative test at this time.
° At week 72, HCV RNA detection should confirm or disprove a sustained virological re-
sponse.
• Patients with cirrhosis should also be referred to a specialist for initial evaluation of their cir-
rhosis.

241
HIV/AIDS TREATMENT AND CARE CLINICAL PROTOCOLS FOR THE WHO EUROPEAN REGION

1.4.2. Algorithm 2
This algorithm is an alternative, focusing on liver biopsy and other tools in the absence of
genotyping.

Fig. 7. Algorithm 2

HIV and HCV positive serology

Child-Pugh B or
HCV RNA + (qualitative or
decompensated
quantitative)
cirrhosis

Liver biopsy or
Mild
non-invasive markersa

Observe

Moderate-severe
Evidence of
clinical & lab
progression Treatment for
12 weeks

HCV RNA
(quantitative)

>2 log
drop or <2 log
negative drop

Continue until Stop


48 weeks treatment

Refer to palliative care

a
FibroScan (image technique), Fibro Test (serum fibromarkers).

242
Management of Hepatitis C and HIV Coinfection

2. Assessment of HIV risk and diagnosis of HIV/AIDS in HCV patients


All patients with HCV should be offered HIV testing and counselling because the two viruses share
transmission routes and because HIV exacerbates the development of HCV. Health-care providers
should explain the reasons for offering the test to patients and its importance in proper clinical man-
agement. However, patients have the right to opt out.
Initial assessment of HIV status should include:
• HIV pretest counselling;
• serological testing (typically enzyme-linked immunosorbent assay (ELISA) and/or rapid tests)
for HIV antibodies, followed by a western blot confirmatory test if positive; and
• post-test counselling, including information on reducing risky behaviour, regardless of whether
the HIV results were positive or negative.

Further clinical evaluation of HIV-infected patients is required to develop a strategy of clinical


management. It should include:
• checking symptoms
• a physical examination
• evaluation of mental health and preparedness for treatment
• a routine laboratory assessment
• a CD4 lymphocyte count to determine the severity of immunodeficiency
• viral load testing if available
• pregnancy testing if indicated
• testing for comorbidities, including hepatitis B, TB and psychiatric disorders
• other tests as indicated by the patient’s condition.

Table 5. Initial and pre-therapeutic evaluation for HCV/HIV-coinfected patients

Pre-
Initial
Tests therapeutic
evaluation
evaluation
HCV disease - qualitative HCV RNA +
- transaminases (ALT, AST), GGT, alkaline phosphatases, +
bilirubin, albumin, prothrombintime
- HCV genotype +
- quantitative HCV RNA +
- ultrasound examination of the liver +
- histological evaluation, non-invasive markers +
HIV a
- CD4 cell count +
- HIV RNA +
- present antiretroviral regimen +
Comorbidities and - HBV serology +
co-conditions - HAV serology +
- TB diagnosis +
- TSH dosage +
- auto-antibodies +
- creatininaemia, proteinuria +
- glycaemia +
- ferritinaemia +
- quantification of alcohol consumption +
- drug consumption +
- pregnancy test +
- ECG (if >50 years old or known cardiopathy) +
- psychiatric consultation if previous psychiatric history +
a
For more information refer to section on initial patient evaluation in Protocol 1, Patient evaluation and antiretroviral treat-
ment for adults and adolescents.

243
HIV/AIDS TREATMENT AND CARE CLINICAL PROTOCOLS FOR THE WHO EUROPEAN REGION

III. Clinical management of HCV/HIV patients

The key issue in the clinical management of HCV/HIV-coinfected patients is the treatment decision
for each condition and when to initiate it. By the end of the laboratory and clinical assessment of
patients with HCV/HIV coinfection, patients can be split into four categories:
1. patients not requiring hepatitis C or HIV/AIDS treatment
2. patients requiring only hepatitis C treatment
3. patients requiring only HIV/AIDS treatment
4. patients requiring both hepatitis C and HIV/AIDS treatment.

1. Coinfected patients not requiring any treatment


Coinfected patients not requiring any treatment meet the following criteria:
• CD4 count >350 cells/mm3 and absence of HIV-related symptoms, and
• HCV antibodies, but absence of HCV RNA replication.
• Coinfected patients not needing treatment should be monitored every six months (clinical fol-
low-up, liver function tests) and every three years for histological liver lesions (using alterna-
tives to liver biopsies).

2. Coinfected patients requiring only HCV treatment


Coinfected patients requiring only HCV treatment meet the following conditions:
• CD4 count >350 cells/mm3 and absence of HIV-related symptoms, and
• active or chronic hepatitis C.

HCV treatment offers the possibility of eradicating HCV within a defined treatment period. In the
following situations, where the benefits outweigh the risks, there are two main reasons to consider
all HCV/HIV-coinfected patients for HCV treatment:
• The liver disease progresses more rapidly to end-stage complications and at earlier ages than in
HCV-monoinfected patients.
• Patients are at higher risk for developing hepatotoxicity following the initiation of ART than
HIV-monoinfected patients. Efficient HCV treatment will hence facilitate the subsequent man-
agement of ART.

2.1. Indications for HCV treatment


• Genotype 2 or 3 regardless of HCV viral load or histology
• Genotype 1, viral load ≤800 000 IU/ml regardless of histology
• Genotype 1 or 4, viral load >800 000 IU/ml and moderate or severe fibrosis

2.2. Predictors of sustained virological response probability


Several baseline parameters can predict a greater likelihood of achieving an SVR (32):
• infection with genotype 2 or 3
• viral load ≤800 000 IU/ml
• absence of cirrhosis
• age <40 years
• ALT levels >3 x upper limit of normal.

7
Some patients may have HCV RNA but harbour genotype 1 or 4 and a mild disease. In such cases, treatment is not recom-
mended; regular yearly monitoring is the recommended option, with an assessment for liver fibrosis after three years.
8
For patients with evidence of advanced liver fibrosis, HCV treatment should be a priority.

244
Management of Hepatitis C and HIV Coinfection

2.3. Contraindications for hepatitis C treatment


The following contraindications for treatment of hepatitis C should be borne in mind:
• pregnancy, because of risk of IFN and RBV9,10
• cardiopathy, such as ischaemic disease and cardiac insufficiency
• psychiatric disorders or history of same
• active alcohol intake (>50 g/day)
• decompensated cirrhosis (Child-Pugh C).11

2.4. Treatment of acute hepatitis C


• Treatment of acute hepatitis C may reduce the risk of chronicity (51). Therefore, if serum HCV
RNA is not eliminated spontaneously within three months of the disease onset (clinically and/or
laboratory documented), treatment with PEG-IFN is recommended for six months (51).
• The use of combination treatment in this population remains a field of research.

2.5. Treatment of chronic hepatitis C (doses and schedules)


All patients should receive a combination of PEG-IFN α2a or α2b and RBV. The standard dose for
PEG-IFN α2a is 180 µg once weekly (QW), and for PEG-IFN α2b it is 1.5 µg/kg body weight QW
(2–5).

The dose of RBV is critical. Although clinical trials in HIV/HCV-coinfected patients have used
a fixed dose of 800 mg per day [400 mg twice daily (BID)] for all genotypes, studies from HCV-
monoinfected patients support the use of 1000 mg to 1200 mg RBV per day (in 2 doses) for treat-
ment of infections with genotypes 1 and 4, and 800 mg RBV per day (400 mg BID) for genotypes
2 and 3 (49).

The current recommendations are as follows:


• for HCV/HIV-coinfected patients with genotype 1 or 4, an initial RBV dose of 1000–1200 mg
once daily (OD);
• for HCV/HIV-coinfected patients with genotype 2 or 3, 800 mg OD (43).

9
Limited data suggest IFN does not have any effect on the embryo or foetus.
10
RBV is teratogenic (causes birth defects) in multiple animal species and its use during pregnancy is contraindicated (48).
Since RBV may cause abnormalities in sperm, men taking it should wait six months after discontinuing use before attempting
to impregnate a woman.
11
IFN is very badly tolerated in these patients (49); however, after regression of the decompensation, treatment may sometimes
be initiated (50) and liver transplantation should be the primary treatment option for such patients.

245
HIV/AIDS TREATMENT AND CARE CLINICAL PROTOCOLS FOR THE WHO EUROPEAN REGION

Fig. 8. Therapeutic algorithm for HCV treatment in HIV-infected patients

Genotype 2 or 3 Genotype 1 or 4

Low HCV RNA High HCV RNA


≤800 000 IU/ml >800 000 IU/ml
<5.9 log IU/ml >5.9 log IU/ml

Ribavirin 800 mg/day Histology evaluation or


non-invasive methods

Ribavirin 1000-1200 mg/day

Regardless of genotype, add either:


PEG-IFN α2a, 180 μg QW
OR
PEG-IFN α2b, 1.5 μg/kg body weight QW

Source: Alberti et al., Sulkowski (43,52)

2.6. Treatment duration


Regardless of genotype, the expected duration of treatment in coinfected patients should be 48
weeks. However, depending on HCV RNA levels at week 12, treatment may be interrupted earlier
(refer to Algorithms 1 and 2 in section II.1.4 above) (43).

Genotype 2 and 3 patients treated for six months have significantly higher relapse rates than those
treated for one year (5, 53). Therefore, all HCV/HIV-coinfected patients should be treated for one
year. HCV genotype can be used as a predictor of response but not as a basis for modifying treat-
ment duration, as with immunocompetent patients.

3. Coinfected patients requiring only HIV/AIDS treatment


Coinfected patients requiring only HIV/AIDS treatment satisfy at least one condition in each of the
following bullet points
• CD4 count ≤350 cells/mm3 in symptomatic patients or patients with viral load >100 000
copies/ml, or CD4 count ≤200 cells/mm3 irrespective of symptoms;
and
• HCV antibodies but no HCV RNA replication, or hepatitis C with contraindications to treatment
(in the knowledge that they may be transient – see section III.2.3 on contraindictions).

3.1. Initiation of HAART


Initiation of ART in HCV/HIV-coinfected patients should follow the current recommendations for
HIV-monoinfected patients (54). (For further details, please refer to Protocol 1, Patient evaluation
and antiretroviral treatment for adults and adolescents.) (see Table 6)

246
Management of Hepatitis C and HIV Coinfection

Table 6. Recommendations for initiating HAART in HCV/HIV-coinfected patients

CD4 cell count Recommendations


CD4 <200 cells/mm3 Antiretroviral treatment
CD4 200–350 cells/ mm3 Antiretroviral treatment should be considered when there is a high viral
or VL > 100 000 copies/ml load, a rapid decline in CD4 count or the presence of symptomatic HIV
disease. It should be started before the CD4 count falls to <200 cells/mm3.

3.2. Considerations in choosing a HAART regimen


In HCV/HIV-coinfected patients, the selection of an adequate first-line regimen should take into
account major concerns and potential problems:
• adherence (a once-daily regimen should be favoured);
• hepatotoxicity of non-nucleoside reverse transcriptase inhibitors (NNRTIs) (acute, such as with
nevirapine (NVP));
• drug interaction: didanosine (ddI) and zidovudine (ZDV) with RBV, efavirenz (EFV) and PEG-
IFN (severe depression);
• use of opioid substitution therapy (OST): pharmacokinetic interaction between NNRTIs and
methadone or buprenorphine (dose adjustments);
• coexistent medical/psychiatric conditions; and
• the same concerns as in monoinfection: potency, maintenance of future options, cost and avail-
ability.

3.3. First-line HAART regimens

Table 7. Treatment Regimens for first-line HAART in HCV/HIV-coinfected patients

ARV drug classes HAART regimens


ZDVa or d4T EFVb
Ó Ï
Preferred first line 2 NRTIs + 1 NNRTI 3TC or FTCc
Ï ¯
NVPb
ABC or TDF

ZDV a
ABCd
Ó Ï
Alternative first line 3 NRTIs 3TC or FTCc
Ï ¯

d4T TDF
a
ZDV is not an absolute contraindication if a patient is on RBV, but haemoglobin (Hb) levels should be closely monitored.
b
EFV has been considered the preferred NNRTI option, but NVP can be considered for patients without evidence of hepatic
dysfunction, with close monitoring. However, it should be avoided in HIV-infected patients if CD4 is >400 cells/mm3 (>250
mm3 in women) (55).
c
Emtricitabine (FTC) is equivalent to 3TC. FTC is available together with TDF, and 3TC together with ABC as fixed-dose
combination (FDCs).
d
ZDV/3TC/ABC regimen is available as an FDC.

• In case of severe toxicity and side-effects in first-line antiretrivorals (ARVs), substituting


another ARV with a different toxicity profile within the front-line regimens is recommended.
• Switching to second-line ARV regimens is recommended in the absence of immunological or
virological response to ART, as measured by CD4 cell count and viral load. (Please refer to Pro-
tocol 1, Patient evaluation and antiretroviral treatment for adults and adolescents for further
details).

247
HIV/AIDS TREATMENT AND CARE CLINICAL PROTOCOLS FOR THE WHO EUROPEAN REGION

3.4. Second-line HAART regimens


For second-line HAART, WHO recommends selecting three different drugs containing at least one
new pharmacological class.
• The best options are regimens with a boosted protease inhibitor (PI) as the key drug, together
with two nucleosides if a classical approach of 2 NRTIs + 1 NNRTI was the first-line treat-
ment.
• In case of a simplified first choice with 3 NRTIs, the second-line should use a boosted PI + 1
NNRTI and/or 1 NRTI.
Among second-line NRTIs, those with better resistant profiles, such as ddI, ABC and TDF, should
be given preference.
• The combination d4T+ddI has to be avoided due to the risk of mitochondrial toxity, leading to
hepatic steatosis and potentially enhancing fibrosis (56).
• TDF/ddi is also contraindicated due to negative pharmacological interactions.

Table 8. Treatment regimens for second-line HAART in HIV/HCV-coinfected patients

ARV drug classes HAART regimens


ABC + TDF LPV/r
or
Preferred second line 2 NRTIs + 1 boosted PI or + SQV/r
or
ABC + ddIa ATZ/rb
ABC Ó Ï LPV/r
1 NNRTI +/- 1 NRTI + 1 boosted PI EFV Ë SQV/r
TDF Ï Ó ATZ/rb
Alternative second line or or
LPV/r + EFV
double PI or
LPV/r + SQV
a
A ddI dose in combination with TDF should be adjusted to less than 4.1 mg/kg per day so as not to compromise immune
recovery. It is contraindicated in patients with cirrhosis and under RBV treatment, and should be used with caution in
patients with less severe liver disease.
b
Unboosted ATZ or NFV can be used in absence of a cold chain.

4. Coinfected patients requiring both HCV and HIV/AIDS treatment


Coinfected patients requiring both HCV and HIV/AIDS treatment meet the following criteria:
• CD4 count ≤350 cells/mm3 in symptomatic patients or patients with viral load >100 000 cop-
ies/ml, or CD4 count ≤200 cells/mm3 irrespective of symptoms; and
• acute or chronic hepatitis C.12

4.1. Strategy for initiation of treatment


See Table 9 below.
• If a coinfected patient has severe immunodeficiency (CD4 count <200 cells/mm3), the CD4
count should be improved using HAART before commencing HCV treatment.
• If CD4 is between 200 and 350 cells/mm3, HCV treatment should be offered first in order to
avoid interactions between HAART and anti-HCV drugs and facilitate adherence. After HCV
treatment is finished (12 months), HAART should be initiated.
• Patients, who need or are receiving HAART, should be in stable treatment (adherence to treat-
ment, absence of side-effects, CD4 >200 cells/mm3) for a few months before starting HCV
treatment. HAART should be continued during HCV treatment but ddI, ZDV or d4T should be
changed for other drugs (ABC, TDF, etc.) before initiating RBV.

12
For patients with evidence of advanced liver fibrosis, HCV treatment should be a priority.

248
Management of Hepatitis C and HIV Coinfection

• In some cases (if CD4 nadir has never been <200 cells/mm3), interruption of HAART during
HCV treatment is feasible if the patient asks for it. In this case, the original regimen is usually
reintroduced after the end of HCV treatment or in case the CD4 count drops <200 cells/mm3
during the treatment.
• Patients with a low baseline CD4 count (<200 cells/mm3) may tolerate HCV treatment less well
and may be at higher risk for developing opportunistic infections, since IFN treatment is often
associated with loss of CD4 cells in the bloodstream, although the CD4 percentage is conserved
(2–5).

Table 9. Algorithm for initiation of hepatitis C treatment and HAART in


HCV/HIV-coinfected patients
Patients HAART HCV treatment

Treat HCV first


No indication for ARV
CD4 >350 cells/mm3

Treat HCV first, then initiate HAART


ARV initiation indicated
CD4 200–350 cells/mm3
Untreated

CD4 <200 cells/mm3 Initiate HAART, wait until stable, and


regimen is well tolerated, then treat HCV
Replace ddI and ZDV if on alternative
options.

ARV-treated It is possible to interrupt HAART until Treat HCV if CD4 > 200 cells/mm3.
the end of HCV treatment (if CD4 nadir
was never <200 cells/mm3, and patient
asks for it).

4.2. Considerations of ARVs when treating both HCV and HIV infections

4.2.1 Zidovudine (ZDV)


ZDV, when taken concomitantly with RBV, is associated with an increased frequency of anaemia,
but not severe neutropenia. When alternative options are available, ZDV should be replaced by
another NRTI during HCV treatment.

4.2.2 Didanosine (ddI)


Didanosine used in association with RBV was shown to be associated with a markedly increased
risk of lactic acidosis, pancreatitis (57, 58) and an unexpected number of hepatic decompensa-
tions in patients with cirrhosis (59). It is consequently contraindicated in patients with cirrhosis
and should be used with caution in patients with less severe liver disease during PEG-IFN + RBV
combination treatment.

4.2.3 Efavirenz (EFV)


EFV and PEG-IFN can be co-prescribed but must not be initiated simultaneously, as both drugs can
induce psychiatric troubles. If EFV is well tolerated then IFN can be added.

4.2.4 Protease inhibitors (PIs)


A potential negative impact of PI use on SVR in patients with HCV/HIV coinfection treated with
PEG-IFN + RBV has been suggested in a subgroup analysis of a single study (25). As there is no
solid evidence regarding this possible negative impact of PI use on SVR, PIs cannot be excluded
from recommended ARVs for HCV/HIV patients. However, more research is needed to obtain
better evidence.

249
HIV/AIDS TREATMENT AND CARE CLINICAL PROTOCOLS FOR THE WHO EUROPEAN REGION

4.3. Hepatotoxicity of ARV drugs


HAART is associated with a higher risk of hepatotoxicity (defined as at least two fold ALT/AST
increase above upper limit of normal (ULN)) in HCV/HIV-coinfected patients than in HIV-monoin-
fected patients (30, 60–64). However, the incidence and risk factors for liver enzyme elevations in
large cohorts of HCV/HIV-coinfected patients are not well defined. In several studies, however,
independent risk factors for hepatotoxicity have been identified (30, 60–64):
• previous liver transaminase elevations to a grade ≥ III
• higher baseline alanine amino-transferase values
• viral coinfection
• high plasma drug levels
• degree of immune damage (64).

Hepatotoxicity has been associated with all currently used ARV drugs, but existing studies fail to
demonstrate a consistent association between particular drugs or drug classes and the development
of subsequent hepatotoxicity. Comparison of HAART regimens (single-PI, multiple-PI and NNRTI-
based) has given inconsistent results for liver-tolerability in cohorts in which HCV/HIV-coinfected
patients are underrepresented.
• Acute hepatotoxicity: in a single cohort study involving HCV positive and negative patients, the
use of NVF within 12 weeks of initiating treatment and the use of full-dose ritonavir (RTV) (600
mg BID) have been implicated (62). But most liver enzyme elevation events are sub-clinical and
usually reverse spontaneously. NVP is not contraindicated in all HCV/HIV-coinfected subjects,
but should be closely monitored when used in asymptomatic patients. A majority of experts
recommend avoiding its use in patients with evidence of liver dysfunction.
• Chronic hepatotoxicity: the prolonged use of nucleoside analogue reverse transcriptase inhibi-
tors (especially of those having a strong affinity for mitochondrial deoxyribonucleic acid (DNA)
polymerase, such as ddI and d4T) exposes treated patients to a risk of chronic mitochondrial
toxicity, whose target, among other organs, is the liver. This toxicity, possibly exacerbated in
some patients by the specific chronic toxicity of PIs on the liver, may lead to hepatic steatosis
and worsen pre-existing fibrosis.

4.4. ARV dose adjustment in patients with cirrhosis


• Like a majority of drugs metabolized in the liver, antiretroviral agents such as PIs and NNRTIs
are metabolized with difficulty in patients with cirrhosis (65, 66).
• Although the relationship between high plasma concentrations and toxicity is not constant for
all antiretroviral agents, it has been clearly demonstrated for certain PIs, such as NFV, LPV and
amprenavir (APV), and NNRTIs such as EFV (67–70).
• Of the NRTIs, only ZDV and ABC are metabolized by liver enzymes other than cytochrome
P450 (CYP) (65, 66). Consequently, use of PIs, NNRTIs, ZDV or ABC in patients with liver-
decompensated cirrhosis requires dosage adjustment in order to avoid a risk of drug accumula-
tion. However, little specific guidance has been established to precisely adapt ARV dosages in
patients with cirrhosis.

250
Management of Hepatitis C and HIV Coinfection

Table 10. Recommendations for antiretroviral dosage adjustment in patients with ESLD

ARV Main metabolism path- Pharmacokinetic in Adjustment recommendation


way ESLD
NRTI
Zidovudine 80% liver glucuronidation Accumulation and in- Dosage adjustment may be useful
and <5% renal elimina- creased risk of haemato- but no specific recommendations.
tion logical toxicity Clinical monitoring and decreased
daily dose in case of intolerance
(anaemia).
Lamivudine 80% renal elimination Not affected No change
Emtricitabine 80% renal elimination No data No change
Stavudine 80% renal elimination Not affected Avoid due to high risk of hepatic
steatosis.
Didanosine 50% renal elimination No data Avoid due to high risk of hepatic
steatosis and pancreatitis.
Tenofovir 80% renal elimination Not affected No change
Abacavir Liver glucuronidation; Accumulation Avoid.
<5% renal elimination
NNRTI
Nevirapine Liver (CYP enzymes) Reduced clearance Avoid due to the risk of severe hepa-
totoxicity (grade 3 or 4).
Efavirenz Liver (CYP enzymes) Reduced clearance Careful monitoring of CNS side-ef-
Little information fects if elevated transaminases.
Drug monitoring if available.
PI
Nelfinavir Liver (CYP enzymes) Reduced clearance Drug monitoring
Indinavir Liver (CYP enzymes) Sparse data Drug monitoring.
If not available, dosage has to be
reduced at least to:
- 600 mg three times daily without
RTV; or
- 600 mg + 100 mg RTV BID.
Saquinavir Liver (CYP enzymes) No data Drug monitoring
Lopinavir/r Liver (CYP enzymes) Altered Drug monitoring
Atazanavir Liver (CYP enzymes) Altered Decrease by 50%.
Amprenavir Liver (CYP enzymes) Altered Decrease the dose:
- to 450 mg BID if Child-Pugh A
- to 300 mg BID if Child-Pugh B–C.
Fosamprenavir Liver (CYP enzymes) Altered Contraindicated if severe liver
disease

Source: Wyles & Gerber, Salmon & Taburet (65, 66).

4.4.1 Recommendations
• In the absence of specific recommendations, the full dose of ARVs is usually prescribed in
patients with compensated cirrhosis.
• If therapeutic drug monitoring is available, residual drug concentrations of ARVs should be
measured at the first monitoring visit in order to adjust dosages.
• In cases of decompensated cirrhosis where drug monitoring is not available, one should:
° avoid NNRTIs
° reduce the daily dosage of ZDV and ABC
° reduce the daily dose of most PIs (precise data are lacking).

251
HIV/AIDS TREATMENT AND CARE CLINICAL PROTOCOLS FOR THE WHO EUROPEAN REGION

5. Clinical monitoring
HCV/HIV coinfected patients should be carefully monitored during treatment. For monitoring of
patients receiving ART please refer to Protocol 1, Patient evaluation and antiretroviral treatment
for adults and adolescents.
Patients treated for HCV should be followed monthly for clinical evaluation of treatment tolerance.
The tests to be regularly performed are shown in Table 11.

Table 11. Monitoring during treatment

Before W4 W8 W12 W16 W20 W24 W28 W32 W34 W36 W48 W72
treat-
ment
Blood count W1
W2
Tolerance

and X X X X X X X X X X X
platelets* W4
CD4 X X X X X X X X X
TSH X X X
Quantitative
HCV viral X X
Efficacy

load
Qualitative
HCV RNA X X X

Note: W=week
* Blood and platelets counts should also occur during weeks 1 and 2.

5.1. Virological response monitoring


See Table 11 above.
The virological response should be monitored by serum HCV RNA quantification before initiation
of treatment and 12 weeks after starting treatment using the same sensitive test with a lower detec-
tion limit of 50 IU/ml:
• For patients with at least a 2 log reduction in viral load at week 12 – defined as an early virologi-
cal response (EVR) – treatment should be continued.
• If a 2 log reduction in viral load is not achieved at week 12, treatment should be stopped, be-
cause the negative predictive value of achieving SVR is 99–100%. This rule is applicable to all
genotypes.

The log rule at week 12 in coinfected patients is of great relevance to optimizing treatment. It
encourages treatment of all candidates in the absence of contraindication, given that treatment can
be stopped after 12 weeks if there is no chance of a cure.

After week 12, assessment should be made by a qualitative HCV RNA test, as follows:
• Week 24: for patients remaining positive for serum HCV RNA at week 24 (negative predictive
value for achieving SVR is 100%), treatment should be discontinued.
• Week 48 marks the end of treatment response.
• Week 72: after six months off treatment, negative HCV RNA indicates an SVR. Recurrence of
HCV infection thereafter is very rare.
• A new assessment might also be useful 12–24 months after the end of treatment.

5.2. Histological response monitoring


A new liver biopsy is not indicated except in patients with no SVR, for whom the result of liver
biopsy could modify HCV treatment.

252
Management of Hepatitis C and HIV Coinfection

5.3. Tolerance monitoring


See Table 11 above.
A full blood count as well as transaminases and bilirubin tests should be performed in weeks 1, 2
and 4, and thereafter on a monthly basis. CD4 cell count should be monitored monthly. Additional
laboratory tests can then be carried out at the physician’s discretion and should include assessment
of thyroid-stimulating hormone (TSH) at least every three months.

5.4. Management of toxicity and side-effects of PEG-IFN + RBV treatment


Side-effects of PEG-IFN and RBV occur in a majority of patients and may be severe (2–5, 71).
Effort should be made to keep patients on the optimal dose of PEG-IFN plus RBV and to proac-
tively manage side-effects of treatment. It is important to maintain the optimal doses of RBV and
PEG-IFN during treatment, especially during the first 12 weeks. The use of erytropoetin may make
it possible to avoid decreasing RBV dosage (72). However, if severe side-effects or laboratory ab-
normalities develop during treatment and no growth factor is available, the dosages of each product
have to be modified until the reactions disappear, as described in section 5.4.2 below.

5.4.1. Anaemia and neutropenia


• Anaemia (<10 g/dl) is reported in up to 30% of patients receiving PEG-IFN + RBV and has been
shown to impair quality of life (2–5, 71).
• Anaemia increases with the concomitant use of ZDV and a lower baseline haemoglobin.
• ZDV should be replaced in patients with ART alternatives.
• Neutropenia (<1000 cells/mm3) is observed in up to 50% of patients, but serious bacterial infec-
tions seem infrequent (2–5, 71).

5.4.2. Dose adjustment of PEG-IFN and RBV

Table 12. Dose adjustment for side-effects and toxicity

Reduce RBV Withhold RBV Reduce PEG- Withhold Discontinue


to 600 mg IFN by 70%, PEG-IFN combination
50%, 25%
Absolute neutrophil <750/mm3 <500/mm3
count
Platelet count 25 000– <25 000/mm3
50 000/mm3
Haemoglobin
- no cardiac disease 8.5–10.0 g/dl <8.5 g/dl

- stable cardiac decrease of <12 g/dl despite


disease ≥2 g/dl during four weeks at
any four weeks reduced dose

Source: European Medicine Agency (73, 74).

• RBV should be reduced to 600 mg/daily (200 mg in the morning and 400 mg in the evening) if
either of the following applies:
° the haemoglobin of a patient without significant cardiovascular disease falls to <10 g/dl and
≥8.5 g/dl; or
° the haemoglobin of a patient with stable cardiovascular disease fall by ≥2 g/dl during any
four weeks of treatment (a return to the original dosage is not recommended).
• RBV should be discontinued if either of the following applies.
° The haemoglobin of a patient without significant cardiovascular disease falls to <8.5 g/dl.
° A patient with stable cardiovascular disease maintains a haemoglobin value <12 g/dl despite
four weeks on a reduced dose.

253
HIV/AIDS TREATMENT AND CARE CLINICAL PROTOCOLS FOR THE WHO EUROPEAN REGION

If the abnormality is reversed, RBV may be restarted at 600 mg daily, and be increased to 800
mg daily at the discretion of the treating physician (a return to the original dosage is not recom-
mended).
• In case of RBV intolerance, PEG-IFN monotreatment should be continued.
• Dose reduction of PEG-IFN is recommended if the neutrophil count is <750/mm3 as described
in Table 12 (53). For patients with an absolute neutrophil count <500/mm3 treatment should be
suspended until values return to >1000/mm3. Treatment should be reinstituted at 50% of the
dose and the neutrophil count monitored.
• A 50% dose reduction is recommended if the platelet count is <50 000/mm3. Cessation of treat-
ment is recommended when platelet count decreases to levels <25 000/mm3.

5.4.3. Influenza-like symptoms


• Paracetamol (possibly combined with non-steroidal anti-inflammatory drugs) should be used
for influenza-like syndrome, particularly before injection of PEG-IFN.
• Low platelets are a relative contraindication for the use of acetylsalicylic acid, diclofenac or
ibuprofen, because of the inhibition of platelet aggregation.
• Dose adjustment may be required in case of severe side-effects despite symptomatic treatment.
An initial dose reduction to 75% or 50% of the dose is generally adequate.

5.4.4. Nausea
Nausea can be reduced with metoclopramide 10 mg three times daily (TID).

5.4.5. Depression
• Depressive mood changes are frequent and should be managed proactively with symptomatic
treatment. In patients with a history of neurotic or minor depression, initiation of treatment with
antidepressants before starting IFN-based treatment should be considered. Antidepressants are
frequently needed for clinically-relevant depression. Use the following dosages:
° selective serotonin reuptake inhibitors such as citaprolamin, paroxetin and tricyclic at initial
dosages of 20 mg/day; and
° antidepressants such as doxepine at an initial dosage of 50 mg/day.
• Consultation with an experienced psychiatrist for the establishment of a standardized treatment
procedure is recommended.
• In patients with pre-existing depressive mood disorders or other profound neurotic disorders,
initiation of specific psychiatric medication is recommended to reduce the destabilizing effect
of IFN-based treatment.
• In patients with a history of hospitalization due to major depression or psychosis, IFN-based
treatment is generally contraindicated. In large controlled studies the incidence of attempted or
completed suicides, psychosis and major depression is <1% (2–5, 71). The choice of treatment
strategy should be made in consultation with a psychiatrist.
• In patients with a history of injecting drug use, benzodiazepines should be avoided because of
their potential to induce addiction.

5.4.6. Dysthyroidism
IFN-induced dysthyroidism occurs in 7% of patients, but does not require treatment interruption.
• Thyroid hormone substitution is used in case of hypothyroidism.
• Beta-blockers are useful to relieve symptoms of hyperthyroidism (75).

5.5. Management of treatment adherence


Even among patients who are appropriate candidates for treatment with IFN, acceptance of treat-
ment is low in HCV/HIV-coinfected populations, predominantly due to treatment side-effects and
toxicity. However, a proportion of patients who initially decline IFN treatment accept it after edu-
cation and peer support programmes to facilitate successful treatment. Patients may continue to

254
Management of Hepatitis C and HIV Coinfection

work if necessary, with possible working time adjustments to accommodate for treatment and drug
reactions.

Counselling is essential to increasing adherence. Physicians should:


• listen to patients’ complaints
• teach them to recognize and manage side-effects
• discuss ways to improve compliance.

A team approach to patient care and management is an effective strategy for increasing adherence.
The team should include physicians, nurses, psychiatrists where relevant and social workers or
other care providers.

Initiatives that have proven effective include directly observed treatment, patient discussion groups,
patient manuals, hotlines and psychological support. For further information on adherence please
refer to Protocol 5, HIV/AIDS treatment and care for injecting drug users, and Protocol 1, Patient
evaluation and antiretroviral treatment for adults and adolescents.

5.6. Management of non-responders


Non-response can be observed in any HCV treatment, ranging from “no viral decline during treat-
ment” to “end-of-treatment virological response and subsequent virological relapse”. The decision
to treat patients again with PEG-IFN plus RBV should be based on:
• type of response
• toleration of the previous treatment
• extent of liver damage
• HCV genotype.

If the therapeutic aim in treating patients with biopsy-proven advanced fibrosis/cirrhosis is to delay
or prevent disease progression in non-responders at week 12 and/or week 24, continuation with
PEG-IFN monotreatment can be considered, since a histological response was observed in about
35% of non-responders who received PEG-IFN + RBV in four pivotal trials (2–5). However, data
on dose, duration and clinical benefits of such maintenance treatment are very scarce in HCV/HIV-
coinfected patients, and further research is needed.

5.7. Management of end-stage liver disease (ESLD)13

5.7.1. Testing for hepatocellular carcinoma (HCC)


Cirrhotic patients should be screened for HCC at four-to-six-month intervals using ultrasonography
and measurement of alfa-fetoprotein levels (43). It has been found that HCC occurs more rapidly
and is more aggressive in patients with HIV infection (76). Patients whose test results are abnormal
should be followed up at referral centres for diagnosis, staging and treatment, which is only avail-
able for early-stage HCC (77).

5.7.2. Testing for oesophageal varices


Annual endoscopy, including the investigation of oesophageal varices in the gastric fundus, is rec-
ommended (43). In the presence of significant oesophageal varices, a prevention of bleeding by
non-cardioselective beta-blockers (associated with variceal ligation in case of > grade 2 varices) is
recommended (78). The most frequently prescribed drug is propanolol at a dosage varying from 40
to 160 mg/day in order to obtain a blocking effect (cardiac pulse reduction of 30%).

13
For further details on ESLD, please see Annex 4.

255
HIV/AIDS TREATMENT AND CARE CLINICAL PROTOCOLS FOR THE WHO EUROPEAN REGION

5.8. Drug–drug interactions

5.8.1. Interactions between HIV drugs and HCV drugs


Interactions of ARV agents and anti-HCV drugs must be taken into account, as they partially ex-
plain the high rate of side-effects in HCV/HIV-coinfected patients treated for HCV.
• RBV competes for phosphorylation with thymidine and cytosine analogues such as ZDV and
d4T (79, 80). However, in controlled trials, no effect of RBV on the efficacy of the ARV com-
bination treatment has been observed (81).
• IFN has a moderate antiretroviral effect which may compensate for the effect of RBV on the
efficacy of the ART regimen (82).
• In contrast, the phosphorylation of ddI is increased by RBV (83–87), which may explain some
side-effects observed in co-administration (56–58).

5.8.2. Interactions among recreational drugs, OST, anti-HCV drugs and ARVs
• No finding of interaction between opioids and anti-HCV drugs has been published.
• All PIs and NNRTIs are substrates and potent inhibitors or inducers of the cytochrome P450 sys-
tem. Many classes of recreational drugs, including benzodiazepines, amphetamines and opioids,
are also metabolized by the liver and can potentially interact with antiretrovirals. Overdoses as a
secondary reaction to interactions between the amphetamine-type stimulants (MDMA) and PIs,
particularly RTV, have been reported.
• ARVs that are CYP3A4 inducers (NVP, EFV and PIs) can decrease the level of methadone,
causing withdrawal symptoms and increasing the risk of relapse into heroin abuse.
• An opiate metabolism can be inhibited or induced by concomitant PIs, so patients should be
monitored for signs of toxicity. Withdrawal symptoms generally occur within 4–10 days of ART
initiation. Withdrawals should be monitored clinically and dose increases of 10 mg increments
from days 8–10 should manage the problem.

5.9. Hepatotoxicity of TB drugs in chronic HCV infection


• The rate of hepatotoxicity is significantly higher in TB patients with HCV or HBV coinfection
(59%) than without coinfection (24%) (88).
• Commonly used anti-TB drugs, such as isoniazid, rifampicin, pyrazinamide and ethambutol, are
all hepatotoxic.
• Pyrazinamide is the most hepatotoxic and should be avoided in TB patients with severe chronic
liver disease (89).
• It is not necessary to adapt doses of anti-TB drugs in hepatic insufficiency.
• In decompensated liver disease, a regimen without pyrazinamide should be used.
• Streptomycin, ethambutol, and a reserve drug such as fluoroquinolone can be used if treatment
is necessary in patients with fulminant liver disease. Consultation by a specialist is required.
• Alternative anti-TB drugs with lower hepatotoxicity (rifabutin, amikacin, ofloxacin, levofloxa-
cin, etc.) might be used in cases of severe liver dysfunction, The treatment of these special cases
should be decided in consultation with an acknowledged expert.
• Hepatotoxicity occurrence justifies a monthly monitoring of liver functions.

256
Management of Hepatitis C and HIV Coinfection

IV. Suggested minimum data to be collected at the clinical


level
The suggested minimum data to be collected is important in the development of key indicators on
access to treatment and its success. Such indicators assist managers in decision-making on ways to
strengthen and expand these services to all those in need.

The following data should be collected at each clinical facility on a regular basis (e.g. monthly,
quarterly or semi-annually):
• number of HIV patients (“seen for care” – this will be the denominator for the data below);
• number of HIV patients coinfected with HCV;
• number of HCV/HIV-coinfected patients with chronic hepatitis C;
• number of HCV/HIV-coinfected patients with chronic hepatitis C receiving:
° only HCV treatment
° only ART
° both treatments; and
• number of HCV/HIV-coinfected patients who have died (in a given period) including cause of
death (e.g. liver-related deaths, HIV/AIDS related mortality or non-HIV/AIDS related mortality
such as accident, overdose or suicide).

257
HIV/AIDS TREATMENT AND CARE CLINICAL PROTOCOLS FOR THE WHO EUROPEAN REGION

Annex 1. Laboratory assays for HCV (31)


Detection of HCV antibodies
Detection of HCV antibodies is the first step in screening patients for suspected HCV infection. Cur-
rently available assays are highly sensitive, and specific HCV antibodies are detected with enzyme im-
munoassays (EIA). These assays detect mixtures of antibodies directed against various HCV epitopes
located in HCV proteins: core, NS3, NS4 and, in third-generation tests, NS5 (1, 6). The specificity and
sensitivity of currently available EIAs for HCV antibodies are greater than 99% in immunocompetent
patients with active viral replication (presence of HCV RNA). For patients with acute HCV infection,
it is important to bear in mind that antibodies may not be detectable for three to eight weeks following
initial infection.

The presence of HCV antibodies is indicative of past or present infection. Antibodies persist indefinitely
in chronically infected patients, but antibody titres may decrease or even disappear in patients who clear
HCV either spontaneously or after ART.

Different types of assays and immunoblot tests, were used in the past to confirm positive EIAs results
in low-risk populations, such as in healthy blood donors. The excellent performance of currently avail-
able EIAs and the general availability of HCV RNA testing make these assays outdated. In blood banks,
nucleic acid testing (NAT) has recently been implemented. With NAT, the presence of HCV RNA is
analyzed in small blood pools and, if a viral genome is detected, an individual analysis of the implicated
blood samples is performed. With the addition of NAT, the risk of HCV transmission has been reduced
to around 1/1 000 000 donations.

Qualitative detection of HCV RNA


HCV RNA can be detected as soon as a few days after infection. In general, qualitative assays to detect
HCV RNA are more sensitive than most currently available quantitative assays. However, the latest
quantitative methods are very sensitive and in the future could become the universally used methods.

The qualitative detection procedure begins with RNA extraction from clinical samples. In most centres
RNA extraction has become fully automated, increasing its reproducibility. Thereafter, the target is am-
plified, either by PCR or TMA.

There are currently two commercially available qualitative assays to detect HCV RNA: one PCR-based
assay (Cobas Amplicor HCV v. 2.0, Roche) with a sensitivity of 50 IU/ml and one TMA assay (Versant
HCV RNA qualitative assay, Bayer) with a sensitivity of 5–10 IU/ml. The specificity of both assays is
close to 100%.

Quantification of HCV RNA


In individuals who become chronically infected, HCV RNA levels are relatively stable over time (90).
HCV RNA quantification can be obtained by two techniques.

1. PCR assays
Quantification is based on amplification of the viral template with a known amount of synthetic RNA
standard added to each reaction. The relative amounts of amplified viral template and standard ampli-
cons are measured at the end of the PCR reaction. More recently, “real time” PCR has been developed,
with many advantages such as simplicity, rapidity, wider linear range of HCV RNA concentrations and
minor risk of contamination. Real-time PCR is already replacing conventional PCR assays.

2. DNA assay
Another approach to quantifying HCV RNA is signal amplification, in which viral genomes are released
from the virions and hybridized in solution using target probes. The HCV RNA with target probes are

258
Management of Hepatitis C and HIV Coinfection

then captured onto microwell plates. Additional target probes bind the viral RNA to branched-DNA
amplifier molecules. The signal is amplified by hybridization of oligonucleotide probes conjugated with
alkaline phosphatase for detection and quantification of the HCV RNA.

Determination of HCV genotype (91)


Two methods can be used to determine HCV genotype:
1. RT-PCR assay, based on analysis of the 5’ untranslated region of the HCV genome is the most com-
monly used method. Typing errors are rare but can occur between genotype 1 and some isolates of geno-
type 4; sub-typing errors might occur in 15–20% of cases. These errors can be explained by the high
degree of nucleotide conservation within this region.

2. Serology: determination of HCV genotype can also be performed by detecting type-specific anti-
bodies. Several antigenic determinants have been identified after epitope mapping of the NS4 and core
proteins of HCV. These epitopes have been used to develop a competitive EIA (Murex HCV EIA) and
an immunoblot assay (RIBA, Chiron Corp).

There are studies demonstrating a lower performance of tests aimed at detecting HCV antibodies in
HIV-infected patients, as well as cases of HCV antibody seroconversion coinciding with the administra-
tion of HAART (probably due to immune restoration). However, latest generation HCV antibody EIAs
have incorporated multiple HCV antigens and are very sensitive in HIV-infected patients. Recently,
sera from 559 HIV-infected and 944 HIV-negative IDUs were tested both for HCV antibodies using a
third-generation assay and for HCV RNA using a commercially available test. Of the HIV-infected indi-
viduals, 547 (97.8%) had detectable HCV antibodies, and only one HCV antibody-negative patient had
detectable HCV RNA (27, 28). The figure was similar for HIV-negative patients, indicating that HCV
antibody screening using latest generation assays is reliable in coinfected patients.

259
HIV/AIDS TREATMENT AND CARE CLINICAL PROTOCOLS FOR THE WHO EUROPEAN REGION

Annex 2. Alternative biochemical tests to assess


hepatic fibrosis
Table 13. Initial reports from all major serum assays
No. of Serum Signifi- Auro Cut-off Sensitiv- Speci- PPVa NPVb Comments
patients markers cant (95% ity ficity
fibrosis CI)
Indirect
assays
Wai et al. 192 APRI Ishak 0.88 ≤ 1.5 41% 95% 88% 64% Simple index;
(92) (AST, plate- ≥3 (0.80– accurately predicts
lets) 0.96) significant fibrosis
and cirrhosis
Forns et 476 Forns Index Metavir 0.86 <4.2 94% 51% 40% 96% Approx. half of
al. (93) (age, GGT, ≥2 those with insig-
choles- nificant fibrosis
terol, platelet detected;. use of
count) cholesterol a con-
founding variable
Ziol at el. 327 FibroScan™ Metavir 0.79 >8.7 56% 91% 88% 56% Excellent for the
(94) (hepatic elas- ≥2 (0.73– detection of cir-
tography) 0.84) rhosis; continuous
variable strength
Imbert- 134 FibroTest™ Metavir 0.87 0.30 87% 59% 63% 85% False positives
Bismut et (α2 macro- ≥2 (SD with inflammation
al. (95) globulin, α2 0.34) and haemolysis;
globulin, large validated
.γglobulin, data reported
apolipopro-
tein A1, GGT
and total
bilirubin)
Castera 183 Combined Metavir 0.88 NA NA NA NA Combined score
et al. FibroScan ≥2 (0.82– appears to enhance
(96) and Fi- 0.92) efficacy
broTest
Direct
assays
Patel et 402 Fibrospect Metavir 0.831 0.36 77% 73% 74% 76% No indeterminate
al. (97) hyaluronic ≥2 score across all
acid, tissue stages
inhibitor
of metal-
loproteinase
1 (TIMP-1)
and alfa2-
macro-glob-
ulin
Kelleher 95 SHASTA Ishak 0.87 0.30 88% 72% 55% 94% Detection of early
et al. (hyaluronic ≥3 fibrosis in HCV/
(98) acid, AST & HIV-coinfected
albumin) patients
Rosen- 1021 ELF Scheuer 0.80 0.102 90.5% 41% 99% 92% Validated for mul-
berg et (Propeptide 3 or 4 (0.76– tiple etiologies;
al. (99) III collagen, 0.85) high reproducibil-
TIMP 1, HA) ity and auto-
mated processing
strength
a
PPV: positive predictive value.
b
NPV: negative predictive value.

260
Management of Hepatitis C and HIV Coinfection

Annex 3. Alcohol screening questionnaires


The following is an overview of the most used and well-established alcohol screening questionnaires.

CAGE Test
CAGE (100) is an acronym of the four questions:
1. Have you ever felt you ought to Cut down on your drinking? (yes/no)
2. Have people Annoyed you by criticizing your drinking? (yes/no)
3. Have you ever felt bad or Guilty about your drinking? (yes/no)
4. Have you ever had a drink first thing in the morning to steady your nerves or get rid of a hangover (Eye-
opener)? (yes/no)

Item responses are scored 0 or 1, with a higher score an indication of alcohol problems. A total score of 2 or
greater is considered clinically significant.

AUDIT Test
The AUDIT Test (101) was developed as a simple method of screening for excessive drinking, alcohol de-
pendence and harmful drinking (see Table 14 below). It has the following advantages:
• cross-national standardization, the only screening test designed for international use;
• identifies hazardous and harmful alcohol use, as well as possible dependence;
• it is brief, rapid and flexible;
• designed for primary health-care workers; and
• focuses on recent alcohol use.

A score of 8 in men and 7 in women indicates a strong likelihood of hazardous or harmful alcohol consump-
tion. A score of 13 or more is suggestive of alcohol-related harm.

261
HIV/AIDS TREATMENT AND CARE CLINICAL PROTOCOLS FOR THE WHO EUROPEAN REGION

Table 14. AUDIT TEST

1. How often do you have a drink containing alcohol?


(4) 4 or more times a
(0) Never (1) Monthly or less (2) 2–4 times a month (3) 2–3 times a week
week
2. How many drinks containing alcohol do you have on a typical day when you are drinking?

(0) 1 or 2 (1) 3 or 4 (2) 5 or 6 (3) 7 to 9 (4) 10 or more

3. How often do you have six or more drinks on one occasion?

(0) Never (1) Less than monthly (2) Monthly (3) Weekly (4) Daily or almost

4. How often during the past year have you found that you were not able to stop drinking once you had started?

(0) Never (1) Less than monthly (2) Monthly (3) Weekly (4) Daily or almost

5. How often during the past year have you failed to do what was normally expected of you because of drinking?

(0) Never (1) Less than monthly (2) Monthly (3) Weekly (4) Daily or almost

6. How often during the past year have you needed a first drink in the morning to get yourself going after a heavy drinking
session?

(0) Never (1) Less than monthly (2) Monthly (3) Weekly (4) Daily or almost

7. How often during the past year have you had a feeling of guilt or remorse after drinking?

(0) Never (1) Less than monthly (2) Monthly (3) Weekly (4) Daily or almost

8. How often during the past year have you been unable to remember what happened the night before because you had
been drinking?

(0) Never (1) Less than monthly (2) Monthly (3) Weekly (4) Daily or almost

9. Have you or has someone else been injured as a result of your drinking?

(0) No (2) Yes, but not in the past year (4) Yes, during the past year

10. Has a relative or friend or a doctor or other health worker been concerned about your drinking or suggested you cut
down?

(0) No (2) Yes, but not in the past year (4) Yes, during the past year

Source: Baber et al. (101).

262
Management of Hepatitis C and HIV Coinfection

Annex 4. Management of end-stage liver disease


Hepatocellular carcinoma
As HIV-infected patients live longer, especially in industrialized countries where they have access to
HAART, HCC may begin to emerge in those who would have otherwise have succumbed to compli-
cations from their primary HIV disease. For this reason, HCC is projected to become an increasingly
significant clinical problem in the HIV populations (76, 102–104).

Early diagnosis of HCC is particularly important in patients coinfected with HCV and HIV, because it
is more aggressive and, in its advanced stages, incurable (59). Prevention, therefore, becomes key to
controlling the health-care burden of this disease.

The recommendations for HCC management developed in 2000 by the European Association for the
Study of the Liver (EASL) (105) are being updated. Such recommendations might be problematic in
view of the wide geographical variations in disease epidemiology and treatment availability. Guidelines
for managing HCC arising in connection with HIV coinfection are lacking.

Early diagnosis
The 2000 EASL guidelines describe patient selection and surveillance intervals (105). Patients with cir-
rhosis should be screened, if liver transplantation is feasible. A screening interval of every six months
has been established to allow detection of tumours <3 cm in diameter. Patients whose screening results
are abnormal should be followed up at referral centres for diagnosis and staging.

Ultrasonography and measurement of alpha-fetoprotein (AFP) levels, at six-month intervals, are the
most commonly used methods to screen patients with cirrhosis for HCC (77, 106). AFP values >400
ng/ml are considered diagnostic of HCC.

Treatment
Treatment for HCC is usually classified as curative or palliative (77, 105). Curative treatment
includes:
• surgical resection
• liver transplantation
• arterial embolization
• percutaneous ethanol injection in patients with small tumours who are not candidates for resec-
tion; a modest survival advantage has been shown for chemoembolization in randomized, con-
trolled trials and one meta-analysis.

Most patients cannot undergo resection or liver transplantation because of underlying cirrhosis or
advanced disease at diagnosis.

Early-stage HCC
A solitary tumour <5 cm, or up to 3 tumours <3 cm, in a patient with well-preserved liver function,
constitutes early-stage HCC (4, 8). Monoinfected patients can be successfully treated with curative
therapies, although response rates and survival benefits are variable. Surgical resection and trans-
plantation yield 5-year survival rates ranging from 60% to 70%. Recurrence, however, can be as
high as 50% at 3 years and 70% at 5 years.

Percutaneous ethanol injection induces a complete response in about 80% of patients whose
tumours are ≤3 cm. Response rates are lower with large or multinodal tumours (105).

263
HIV/AIDS TREATMENT AND CARE CLINICAL PROTOCOLS FOR THE WHO EUROPEAN REGION

Advanced HCC
Most patients with HCC (approximately 50%) have advanced disease at diagnosis (77, 105). Pa-
tients with advanced disease are candidates for loco-regional or systemic treatments rather than
curative approaches (4). Transarterial chemoembolization is the only palliative therapy that has
been shown to improve survival, with careful patient selection.

Prevention and recurrence


HIV patients are likely to have other risk factors predisposing them to HCC, such as alcohol abuse
and concurrent HBV infection. Among HIV patients, vaccination against HBV is strongly recom-
mended. HCV/HIV-coinfected patients should receive treatment for chronic HCV infection using
combination IFN and RBV.

Orthotopic liver transplantation


Orthotopic liver transplantation (OLT), where available, is the only therapeutic option for patients
with end-stage liver disease. Accumulated experience in North America and Europe in the last five
years indicates that three-year survival in selected HIV-infected recipients of liver transplants was
similar to that of HIV-negative recipients (107–110). HIV infection by itself is not, therefore, a con-
traindication for liver transplantation.

As the survival of HIV-infected patients with ESLD is shorter than that of non-HIV-infected pa-
tients, the OLT evaluation should be done after the first liver decompensation. The current selection
criteria for HIV-positive transplant candidates include:
• no history of opportunistic infections or HIV-related neoplasms, except infections that can be
efficaciously treated and prevented, such as TB, candidiasis or Pneumocystis jirovecii pneumo-
nia (PCP);
• CD4 cell count >100 cells/mm3; and
• plasma HIV viral load suppressible with antiretroviral treatment.

For drug users, a two-year abstinence from heroin and cocaine is also required, although patients in
a methadone programme can be accepted.

The main problems in the post-transplant period are pharmacokinetic and pharmacodynamic in-
teractions between ARVs and immunosuppressors, and the management of HCV infection relapse,
one of the main causes of post-transplant mortality. Experience with PEG-IFN and RBV is scarce
in this population.

Table 15. Three-year survival of patients with and without HIV-infection who had a
liver transplant before and during the HAART period
Before HAART During HAART period
(<1996) (1996–2004)
Survival
HIV-infected patients HIV-infected patients Non-HIV-infected patients
(n = 32) (n = 24) (UNOS) (n = 5225)
One year 69% 87% 87%
Two years 56% 73% 82%
Three years 44% 73% 79%

Source: Tzakis et al., Miró et al., Ragni et al., (108–110).

264
Management of Hepatitis C and HIV Coinfection

Annex 5. Research needs and alternative treatments


Epidemiology
Studies on the epidemiology and the social impact of HCV in patients infected with HIV should be
actively investigated, with a special emphasis on vulnerable populations.

HIV management
Studies addressing the optimal time in the course of chronic HIV infection to commence ART in
HCV-coinfected patients should be initiated.

HCV management and physiopathology


• Studies to validate the utility of non-invasive methods of liver disease progression should be
performed.
• Long-term follow-up studies of patients with and without SVR are strongly encouraged to deter-
mine late relapses, the duration of histological improvement and the effect of clinically relevant
outcomes such as decompensation, HCC and death.
• Studies on pathophysiology, including extrahepatic viral reservoirs and the specific immune
response to HCV, should be conducted.

Future directions for treatment


Research should also investigate:
• optimizing the response to existing treatments, such as higher doses of RBV or PEG-IFN
• treatment durations
• the utility of maintenance treatment
• the optimal regimen for delaying disease progression.

Higher doses of RBV


The optimal RBV dose for treatment of HCV genotype 1 and the potential benefits of prolonged
treatment should be investigated. The optimal dose of RBV remains unclear. It is possible that
higher SVR rates can be achieved by higher doses of RBV. In most of the published literature on
HIV/HCV-coinfected patients, the RBV dose was 800 mg, in order to avoid anaemia, which was
considered a greater problem in HIV-infected patients, especially those taking ZDV. However, in
HIV-negative patients with genotype 1 HCV infection, it is clear that higher SVR rates are achieved
with 1.0/1.2 g RBV (≤75 kg/>75 kg) than with 800 mg (49). Thus, alternative strategies for HIV-
infected patients need also to consider higher RBV doses. It is important to note that higher RBV
doses appeared to be well tolerated in the Barcelona study (5), where RBV was given by body
weight as follows (per day): 800 mg, <60 kg; 1 g, 60–75 kg; and 1.2 g, >75 kg.

Higher doses of IFN


It is possible that higher SVR rates can be achieved by higher doses of IFN but this has not been
investigated in HIV-infected patients.

Treatment duration
A shorter duration of treatment for patients with HCV genotypes 2 and 3 should be investigated.

In HIV-negative patients, SVR rates are the same for genotype 2 and 3 HCV infections if they are
treated for 24 weeks instead of 48. However, analogous studies have not been reported for PLHIV
(5). Thus, studies emphasizing alternative dosing intervals are also needed for genotype 2 and 3
HCV infection before shorter regimens can be recommended. On the other hand, it might be useful
to evaluate the usefulness of longer treatment duration for genotype 1 HCV infections with high
viral loads.

265
HIV/AIDS TREATMENT AND CARE CLINICAL PROTOCOLS FOR THE WHO EUROPEAN REGION

IFN maintenance treatment


Studies on the use of maintenance treatment in patients with no SVR and with advanced liver disease
are strongly recommended, including evaluation of the optimal dose and duration of treatment.
Maintenance treatment is aimed at decreasing the incidence of ESLD without effecting SVR. The
histologic response results of the ACTG 5071 study (4) described above provide a rationale for this
approach. There are studies designed to test this hypothesis in both HIV-infected (SLAM-C) and
HIV-uninfected people (HALT-C), but it remains undecided.

Acute HCV infection


The optimal treatment for acute HCV infection in HIV-infected patients should be investigated.

New treatments
As the current therapies are suboptimal in efficacy, tolerability and quality of life, the development
of new drugs to improve these issues should be actively pursued.

Phase II and III trials of new drugs should be performed in HIV/HCV-coinfected patients as a prior-
ity due to the accelerated course of hepatitis infections in these populations.

There are many compounds under development, and some have progressed into Phase II clinical
studies (111):
• Viramidine (Valeant) is a prodrug of RBV that causes substantially less anaemia. In phase II
studies, it was associated with less anaemia than RBV and SVR rates that were not inferior.
Phase III studies are underway.
• Albuferon-alfa™ (Human Genome Sciences), is a fusion of albumin and IFN that prolongs IFN
half-life.
• Interleukine-2 (IL-2) treatment has also been examined as a method to boost HCV antibody im-
mune responses and enhance treatment responses. However, an early study in HIV/HCV-coin-
fected patients was associated with significant toxicity and provided no evidence of effective-
ness (10).
• NM283 (Idenix) interferes with the HCV polymerase and, in Phase II studies; its use was associ-
ated with a modest reduction in HCV RNA levels.
• VX 950 (Vertex) is an HCV protease inhibitor that is being examined in clinical trials.

The development of direct antivirals that block essential viral enzymes represents a straightfor-
ward approach to developing new agents to target HCV. Although all HCV enzymes are, in theory,
equally appropriate for therapeutic intervention, the NS3–4A serine protease and the NS5B RNA
polymerase have emerged as the most popular targets. A number of competitive inhibitors of the
NS3 protease as well as nucleoside and non-nucleoside inhibitors of the NS5B polymerase are be-
ing developed. The efficacy shown by NS3 serine protease and the NS5B RNA-dependent RNA
polymerase inhibitors in recent proof-of-concept clinical trials has validated the effort of finding
clinical candidates and triggered a renewed interest in this area (112).

266
Management of Hepatitis C and HIV Coinfection

Table 16. A sample of the drug pipeline for hepatitis C

Compound Company Clinical phase Target Mechanism of action

BILN 2061 (Ciluprevir) Boehringer-Ingelheim Phase II a


NS3–4A protease Product-derived serine prote-
ase inhibitor
VX-950 Vertex/Mitsubishi Phase Ib NS3–4A protease Serine protease reversible
covalent inhibitor
NM283 (Valopici- Idenix/Novartis Phase II NS5B polymerase Nucleoside analogue (chain
tabine) terminator)
JTK-103 Japan Tobacco Phase II NS5B polymerase Non-nucleoside allosteric
inhibitor
HCV-796 ViroPharma/Wyeth Phase Ia NS5B polymerase Non-nucleoside allosteric
inhibitor
Host targets/immunomodulators
Actilon (CpG-10101) Coley Pharmaceutical Phase Ib Toll-like receptor-9 Immunomodulator
Group
ANA245 (Isatoribine) Anadys Pharmaceu- Phase Ib Toll-like receptor-7 Immunomodulator
ticals
ANA975 Anadys Pharmaceu- Phase Ia Toll-like receptor-7 Immunomodulator (prodrug
ticals of ANA245)
a
Development has been halted due to cardiotoxicity in monkeys.
Source: Nunes et al. (42).

267
HIV/AIDS TREATMENT AND CARE CLINICAL PROTOCOLS FOR THE WHO EUROPEAN REGION

References
1. Salmon-Ceron D et al. Liver disease as a major cause of death among HIV-infected patients: roles of
hepatitis C and B viruses and alcohol. Journal of Hepatology, 2005, 42: 799–805.
2. Carrat F et al. Pegylated interferon alfa-2b vs standard interferon alfa-2b, plus ribavirin, for chronic
hepatitis C in HIV-infected patients: a randomized controlled trial. JAMA, 2004, 292:2839–2848.
3. Torriani FJ et al. Peginterferon Alfa-2a plus ribavirin for chronic hepatitis C virus infection in HIV-in-
fected patients. The New England Journal of Medicine, 2004, 351:438–450.
4. Chung RT et al. Peginterferon Alfa-2a plus ribavirin versus interferon alfa-2a plus ribavirin for chronic
hepatitis C in HIV-coinfected persons. The New England Journal of Medicine, 2004, 351:451–459.
5. Laguno M et al. Peginterferon alfa-2b plus ribavirin compared with interferon alfa-2b plus ribavirin for
treatment of HIV/HCV coinfected patients. AIDS, 2004, 18:F27–F36.
6. Rockstroh JK et al. Influence of hepatitis C virus infection on HIV-1 disease progression and response
to highly active antiretroviral therapy. Journal of Infectious Diseases, 2005, 15, 192(6):992–1002.
7. Rockstroh JK et al. F12/4: influence of hepatitis C coinfection on HIV disease progression within the
EUROSIDA Cohort. Ninth European AIDS Conference (EACS): 1st EACS Resistance and Pharmacol-
ogy Workshop, Warsaw, 25–29 October 2003.
8. Sherman KE et al. Prevalence of antibodies to hepatitis C virus in patients infected with the human im-
munodeficiency virus. Journal of Infectious Diseases, 1991, 163:414–415.
9. Salmon-Céron et al. Hospitalized HIV-HCV coinfected patients. A French national survey made in June
2001. Médecine et maladies infectieuses, 2003, 33:78–83.
10. Saillour F et al. Prevalence and determinants of antibodies to hepatitis C virus and mark-
ers for hepatitis B virus infection in patients with HIV infection in Aquitaine. BMJ, 1996, 313:
461–464.
11. Hayashi PH et al. Prevalence of hepatitis C virus antibodies among patients infected with
human immunodeficiency virus. Journal of Medical Virology, 1991, 33: 177–180.
12. Sulkowski MS, Thomas DL. Hepatitis C in the HIV-infected patient. Clinical Liver Disease, 2003,
7(1):179–194.
13. Alter MJ. Epidemiology of viral hepatitis. Journal of Hepatology, 2006, 44(S1):S6–S9.
14. Quaglio GL et al. Hepatitis C virus infection: prevalence, predictor variables and prevention opportuni-
ties among drug users in Italy. Journal of Viral Hepatitis, 2003, 10(5):394–400.
15. D’Oliveira A Jr et al. Prevalence and sexual risk of hepatitis C virus infection when human immunodefi-
ciency virus was acquired through sexual intercourse among patients of the Lyon University Hospitals,
France, 1992–2002. Journal of Viral Hepatitis, 2005, 12(3):330–332.
16. Chaix M-L et al. Homosexually transmitted HCV acute infection related to a clustered genotype 4
HCV in HIV-1-infected men and inefficacy of early antiviral therapy. In: Program and abstracts of the
12th Conference on Retroviruses and Opportunistic Infections. Boston, 22–25 February 2005 (Abstract
122).
17. Ackerman Z, Ackerman E, Paltiel O. Interfamilial transmission of hepatitis C virus: a systematic re-
view. Journal of Viral Hepatology, 2000, 7(2):93–103.
18. Jager J et al., eds. Hepatitis C and injecting drug use: impact, costs and policy options. Lisbon, Euro-
pean Monitoring Centre for Drugs and Drug Addiction, 2004 (EMCDDA Monographs).
19. Franciscus A. HCV Genotype and quasi-species. HCSPFACT Sheet. Hepatitis C Support Project, 2006
(http://www.hcvadvocate.org/hepatitis/factsheets_pdf/genotype_FS.pdf, accessed 28 February 2006).
20. Simmonds et al. Epidemiological, clinical and therapeutic associations of hepatitis C types in western
European patients. Journal of Hepatology, 1996, 24(5):517–524.
21. Zeuzem S et al. Risk factors for the transmission of hepatitis C. Journal of Hepatology, 1996, 24(2
Suppl.):3–10.
22. Salmon D et al. Therapeutic management of hepatitis and HIV infection in coinfected patients: results
of a survey performed before the 2005 Consensus Conference. Journal of Hepatology, 2006, 44(S1):
S2–S5.
23. Poynard T et al. A comparison of fibrosis progression in chronic liver diseases. Journal of Hepatology,
2003, 38:257–265.

268
Management of Hepatitis C and HIV Coinfection

24. Grebely J et al. Effect of HIV coinfection on spontaneous clearance of hepatitis C virus (HCV) in the
downtown Eastside of Vancouver. 3rd International AIDS Society Conference on HIV Pathogenesis and
Treatment, Rio de Janeiro, 24–27 July, 2005 (Abstract No. TuPe1.1C18).
25. Vallet-Pichard A, Pol S. Natural history and predictors of severity of chronic hepatitis C virus (HCV) and
human immunodeficiency virus (HIV) coinfection. Journal of Hepatology, 2006, 44(S1):S28–S34.
26. Benhamou Y et al. Liver fibrosis progression in HIV-HCV coinfected patients. The Multivirc Group.
Hepatology, 1999, 30:1054–1058.
27. Forns X, Costa J. HCV virological assessment. Journal of Hepatology, 2006, 44(S1):
S40–S43.
28. Thio CL et al. Screening for hepatitis C virus in human immunodeficiency virus-infected individuals.
Journal of Clinical Microbiology, 2000, 38(2):575–577.
29. Van Asten L, Prins M. Infection with concurrent multiple hepatitis C virus genotypes is associated with
faster HIV disease progression. AIDS, 2004, 18(17):2319–2324.
30. Nunez M, Soriano V. Hepatotoxicity of antiretrovirals: incidence, mechanisms and management. Drug
Safety, 2005, 28(1):53–66.
31. Pawlotsky JM. Use and interpretation of virological tests for hepatitis C. Hepatology, 2002, 36(5 Suppl.
1):S65–S73.
32. Thomas D. Options for treatment of hepatitis C in HIV-infected persons. Journal of Hepatology, 2006,
44(Suppl. 1):S40–S43.
33. Fried MW et al. Peginterferon alfa-2a plus ribavirin for chronic hepatitis C virus infection. The New
England Journal of Medicine, 2002, 347(13):975–982.
34. Torriani FJ et al. Peginterferon Alfa-2a plus ribavirin for chronic hepatitis C virus infection in HIV-in-
fected patients. The New England Journal of Medicine, 2004, 351(5):438–450.
35. Leruez-Ville M et al. Large-scale analysis of hepatitis C virus serological typing assay: effectiveness
and limits. Journal of Medical Virology, 1998, 55(1):18–23.
36. Pugh RNH et al. Preoperative assessment of patients with liver disease. British Journal of Surgery,
1973, 60:646–649.
37. Bravo AA, Sheth SG, Chopra S. Liver biopsy. The New England Journal of Medicine, 2001, 344(7):495–
500.
38. Friedman SL. Score Metavir Evaluation of fibrosis and hepatitis C. American Journal of Medicine,
1999, 107(6B):27S–30S.
39. Kelleher TB, Afdha NL. Assessment of liver fibrosis in coinfected patients. Journal of Hepatology,
2006, 44(S1):S126–S131.
40. Nunes D et al. HIV infection does not affect the performance of non-invasive markers of
fibrosis for the diagnosis of hepatitis C virus-related liver disease. Journal of Acquired Immune Defi-
ciency Syndrome, 2005, 4(5):538–544).
41. Ce Ledinghen V et al. Diagnosis of hepatic fibrosis and cirrhosis by transient elastography in HIV/hepa-
titis C virus-coinfected patients. Journal of Acquired Immune Deficiency Syndrome, 2006, 41(2):175–
179.
42. Nunes D et al. HIV infection does not affect the performance of non-invasive markers of
fibrosis for the diagnosis of hepatitis C virus-related liver disease. Journal of Acquired Immune Defi-
ciency Syndrome, 2005, 40(5):538–544.
43. Alberti A et al. Short statement of the first European Consensus Conference on the Treatment of Chron-
ic Hepatitis B and C in HIV Coinfected Patients. Journal of Hepatology, 2005, 42(5):615–624.
44. Hassan MM. Risk factors for hepatocellular carcinoma: synergism of alcohol with viral hepatitis and
diabetes mellitus. Hepatology, 2002, 36:1206–1213.
45. Samet JH et al. A randomized controlled trial to enhance antiretroviral therapy adherence in patients
with a history of alcohol problems. Antiviral Therapy, 2005, 10(1):83–93.
46. European STD Guidelines. International Journal of STD & AIDS, 2001, 12(10) Supplement 3.
47. Mast EE et al. Risk factors for perinatal transmission of hepatitis C virus (HCV) and the natural history
of HCV infection acquired in infancy. Journal of Infectous Diseases, 2005, 192(11):1880–1889.
48. Kochhar DM, Penner JD, Knudsen TB. Embryotoxic, teratogenic, and metabolic effects of ribavirin in
mice. Toxicology and Applied Pharmacology, 1980, 52(1):99–112.
49. Hadziyannis SJ et al. Peginterferon-alpha2a and ribavirin combination therapy in chronic hepatitis
C: a randomized study of treatment duration and ribavirin dose. Annals of Internal Medicine, 2004,
140(5):346–355.

269
HIV/AIDS TREATMENT AND CARE CLINICAL PROTOCOLS FOR THE WHO EUROPEAN REGION

50. Marrache F et al. Safety and efficacy of peginterferon plus ribavirin in patients with chron-
ic hepatitis C and bridging fibrosis or cirrhosis. Journal of Viral Hepatology, 2005, 12(4):
421–428.
51. Vogel M et al. Treatment of acute hepatitis C infection in HIV-infected patients: a retrospective analysis
of eleven cases. Journal of Viral Hepatology, 2005, 12(2):207–211.
52. Sulkowski MS. Treatment algorithm for the management of hepatitis C in HIV-coinfected per-
sons. Journal of Hepatology, 2006, 44(Suppl.):S49–S55 (http://www.jhep-elsevier.com/article/PI-
ISO168827500735X/fulltext#, accessed 30 March 2006).
53. Perez-Olmeda M et al. Pegylated IFN-alpha2b plus ribavirin as therapy for chronic hepatitis C in HIV-
infected patients. AIDS, 2003, 17(7):1023–1028.
54. Scaling up antiretroviral therapy in resource-limited settings: treatment guidelines for a public health
approach: 2003 revision. Geneva, World Health Organization, 2004.
55. Patel SM et al. Serious adverse cutaneous and hepatic toxicities associated with nevirapine use by non-
HIV-infected individuals. Journal of Acquired Immune Deficiency Syndrome, 2004, 35(2):120–125.
56. Moreno A et al. High rate of didanosine-related mitochondrial toxicity in HIV-HCV coinfected patients
receiving didanosine. Antiviral Therapy, 2004, 9:133–138.
57. Salmon-Céron D et al. Mitochondrial toxic effects of ribavirin. The Lancet, 2001, 357:1803.
58. Lafeuillade A, Hittinger G, Chapadaud S. Increased mitochondrial toxicity with ribavirin in HIV-HCV
coinfection. The Lancet 2001, 357:280–281.
59. Mauss S. Risk factors for hepatic decompensation in patients with HIV/HCV coinfection and liver cir-
rhosis during interferon-based therapy. AIDS, 2004, 18(13):F21–25.
60. Rodriguez-Rosado R, Garcia-Samaniego J, Soriano V. Hepatotoxicity after introduction of highly ac-
tive antiretroviral therapy. AIDS, 1998, 12:1256.
61. Sulkowski MS et al. Hepatotoxicity associated with antiretroviral therapy in adults infected with human
immunodeficiency virus and the role of hepatitis C or B virus infection. JAMA, 2000, 283:74–80.
62. Wit FW et al. Incidence of and risk factors for severe hepatotoxicity associated with antiretroviral com-
bination therapy. Journal of Infectious Diseases, 2002, 186:23–31.
63. Aceti A et al. Hepatotoxicity development during antiretroviral therapy containing protease
inhibitors in patients with HIV: the role of hepatitis B and C virus infection. Journal of Acquired Im-
mune Deficiency Syndrome, 2002, 29:41–48.
64. Torti C et al. Incidence and risk factors for liver enzyme elevation during highly active antiretroviral
therapy in HIV-HCV coinfected patients: results from the Italian EPOKA-MASTER Cohort. BMC In-
fectious Diseases, 2005, 5:58.
65. Wyles DL, Gerber JG. Antiretroviral drug pharmacokinetic in hepatitis with hepatic dysfunction. Clini-
cal Infectious Diseases, 2005, 40:174–181.
66. Salmon D, Taburet AM. Antiretroviral agents in HIV-infected patients with cirrhosis. Actuality on HIV
in 2005. La Presse médicale, 2005, 34, 10(Suppl. 1):S451–S52, 45.
67. Regazzi M et al. Clinical pharmacokinetics of nelfinavir and its metabolite M8 in human
immunodeficiency virus (HIV)-positive and HIV-hepatitis C virus-coinfected subjects. Antimicrobial
Agents and Chemotherapy, 2005, 49(2):643–649.
68. Arribas JR et al. Lopinavir/Ritonavir as single-drug therapy for maintenance of HIV-1 viral suppres-
sion: 48-week results of a randomized, controlled, open-label, proof-of-concept pilot clinical trial (OK
study). Journal of Acquired Immune Deficiency Syndrome, 2005, 40(3):280–287.
69. Veronèse L et al. Single-dose pharmacokinetics of Amprenavir, a human Immunodeficiency Virus Type
1 protease inhibitor in subjects with normal or impaired hepatic function. Antimicrobial Agents and
Chemotherapy, 2002, 821–826.
70. Dominguez S et al. The HEPADOSE Study: evaluation of protease inhibitors and non-nucleoside
analogue plasma concentrations in HIV/HCV and HIV-infected patients. 3rd International AIDS So-
ciety Conference on HIV Pathogenesis and Treatment, Rio Janeiro, 24–27 July 2005 (Abstract No.
WePp0305; http://www.aegis.com/conferences/IASHIVPT/2005/WePp0305.pdf, accessed 28 February
2006).
71. Chutaputti A. Adverse effects and other safety aspects of the hepatitis C antivirals. Journal of Gastro-
enterology and Hepatology, 2000, 15(Suppl.E):156–163.
72. Sulkowski MS et al. Epoetin alfa once weekly improves anaemia in HIV/hepatitis C virus-coinfected
patients treated with interferon/ribavirin: a randomized controlled trial. Journal of Acquired Immune
Deficiency Syndrome, 2005, 39(4):504–506.

270
Management of Hepatitis C and HIV Coinfection

73. European Medicine Agency. Dosage adjustment of ribavirin Rebetol. London, 2006 (http://www.emea.
eu.int/humandocs/PDFs/EPAR/Rebetol/H-246-PI-en.pdf, accessed 28 February 2006).
74. European Medicine Agency. Dosage adjustment interferon Pegasys and Viraferon Peg. London, 2006
(http://www.emea.eu.int/humandocs/PDFs/EPAR/pegasys/H–395–PI–en.pdf and http://www.emea.
eu.int/humandocs/PDFs/EPAR/Viraferonpeg/H–329–PI–en.pdf, accessed 28 February 2006).
75. Moncoucy X et al. Risk factors and long-term course of thyroid dysfunction during antiviral treatments
in 221 patients with chronic hepatitis C. Gastroenterology and Clinical Biology, 2005, 29(4):339–345.
76. Puoti M et al. Hepatocellular carcinoma in HIV-infected patients: epidemiological features, clinical
presentation and outcome. AIDS, 2004, 18(17):1–9.
77. Hoofnagle JH. Hepatocellular carcinoma: summary and recommendations. Gastroenterology, 2004,
127:S319–S323.
78. Samonakis DN et al. Management of portal hypertension. Postgraduate Medical Journal, 2004,
80(949):634–641.
79. Vogt MW et al. Ribavirin antagonizes the effect of azidothymidine on HIV replication.
Science, 1987, 235:1376–1379.
80. Sim SM et al. Effect of ribavirin on zidovudine efficacy and toxicity in vitro: a concentration-dependent
interaction. AIDS Research and Human Retroviruses, 1998, 14:1661–1667.
81. Salmon-Céron D et al. Interferon-ribavirin in association with stavudine has no impact on plasma hu-
man immunodeficiency virus (HIV) type 1 level in patients coinfected with HIV and hepatitis C virus:
a CORIST–ANRS HC1 trial. Clinical Infectious Diseases, 2003, 36:1295–1304.
82. Perronne C. Antiviral hepatitis and antiretroviral drug interactions. Journal of Hepatology, 2006, 44(S1):
S119–S125.
83. Baba M et al. Ribavirin antagonizes inhibitory effects of pyrimidine 2’,3’–dideoxynucleosides but en-
hances inhibitory effects of purine 2’, 3’– dideoxynucleosides on replication of human immunodefi-
ciency virus in vitro. Antimicrobial Agents and Chemotherapy, 1987, 31:1613–1617.
84. Hoggard PG, et al. Effects of drugs on 2’,3’–dideoxy–2’,3’–didehydrothymidine phosphorylation in
vitro. Antimicrobial Agents and Chemotherapy, 1997, 41:1231–1236.
85. Balzarini J et al. Mechanisms of the potentiating effect of ribavirin on the activity of 2’,3’–
dideoxyinosine against human immunodeficiency virus. Journal of Biological Chemistry, 1991,
266:21:509–514.
86. Harvie P et al. Ribavirin potentiates the efficacy and toxicity of 2’,3’–dideoxyinosine in the murine ac-
quired immunodeficiency syndrome model. Journal of Pharmacology and Experimental Therapeutics,
1996, 279:1009–1017.
87. Japour AJ et al. A phase-1 study of the safety, pharmacokinetics, and antiviral activity of
combination of didanosine and ribavirin in patients with HIV-1 disease. Journal of Acquired Immune
Deficiency Syndrome, 1996, 13:235–246.
88. Ungo JR et al. Antituberculosis drug-induced hepatotoxicity. The role of hepatitis C virus and the hu-
man immunodeficiency virus. American Journal of Respiratory and Critical Care Medicine, 1998,
157(6 Pt 1):1871–1876.
89. Yee D et al. Incidence of serious side-effects from first-line antituberculosis drugs among patients
treated for active tuberculosis. American Journal of Respiratory and Critical Care Medicine, 2003,
167(11):1472–1477.
90. Hollingsworth RC et al. Serum HCV RNA levels assessed by quantitative NASBA: stability of viral
load over time, and lack of correlation with liver disease. The Trent HCV Study Group. Journal of
Hepatology, 1996, 25(3):301–306.
91. Forns X, Bukh J. Methods for determining the hepatitis C virus genotype. Viral Hepatitis
Reviews, 1998, 4:1–19.
92. Wai CT et al. A simple non-invasive index can predict both significant fibrosis and cirrhosis in patients
with chronic hepatitis C. Hepatology, 2003, 38(2):518–526.
93. Forns X et al. Identification of chronic hepatitis C patients without hepatic fibrosis by a simple predic-
tive model. Hepatology, 2002, 36(4 Pt 1):986–992.
94. Ziol M et al. Non-invasive assessment of liver fibrosis by measurement of stiffness in patients with
chronic hepatitis C. Hepatology, 2005, 41(1):48–54.
95. Imbert-Bismut F et al. Biochemical markers of liver fibrosis in patients with hepatitis C virus infection:
a prospective study. The Lancet, 2001, 357(9262):1069–1075.

271
HIV/AIDS TREATMENT AND CARE CLINICAL PROTOCOLS FOR THE WHO EUROPEAN REGION

96. Castera L et al. Prospective comparison of transient elastography, Fibrotest, APRI and liv-
er biopsy for the assessment of fibrosis in chronic hepatitis C. Gastroenterology, 2005, 128(2):
343–350.
97. Patel K et al. Evaluation of a panel of non-invasive serum markers to differentiate mild from moder-
ate-to-advanced liver fibrosis in chronic hepatitis C patients. Journal of Hepatology, 2004, 41(6):935–
942.
98. Kelleher TB et al. Prediction of hepatic fibrosis in HIV/HCV coinfected patients using serum fibrosis
markers: the SHASTA index. Journal of Hepatology, 2005, 43(1):78–84.
99. Rosenberg WM et al. Serum markers detect the presence of liver fibrosis: a cohort study. Gastroenterol-
ogy, 2004, 127(6):1704–1713.
100. Ewing JA. Detecting alcoholism: the CAGE questionnaire. JAMA, Journal of the American Medical
Association, 1984, 252:1905–1907.
101. Babor TF et al. AUDIT, the Alcohol Use Disorders Identification Test: guidelines for use in primary
care (2nd ed.). Geneva, World Health Organization, 2001 (http://whqlibdoc.who.int/hq/2001/WHO_
MSD_MSB_01.6a.pdf, accessed 29 March 2006).
102. Smukler AJ, Ratner L. Hepatitis viruses and hepatocellular carcinoma in HIV-infected
patients. Current Opinion in Oncology, 2002, 14:538–542.
103. Garcia-Samaniego J et al. Hepatocellular carcinoma in HIV-infected patients with chronic hepatitis C.
American Journal of Gastroenterology, 2001, 96:179–183.
104. Davila JA et al. Hepatitis C infection and the increasing incidence of hepatocellular carcinoma: a popu-
lation-based study. Gastroenterology, 2004, 127:1372–1380.
105. Bruix J et al. Clinical management of hepatocellular carcinoma. Conclusions of the Barcelona-2000
EASL conference. European Association for the Study of the Liver. Journal of Hepatology, 2001,
3:421–430.
106. Daniele B et al. alpha-fetoprotein and ultrasonography screening for hepatocellular carcinoma. Gastro-
enterology, 2004, 127:S108–S112.
107. Samuel D et al. Liver transplantation in patients with HIV infection. Journal of Hepatology, 2003,
39(1):3–6.
108. Tzakis AG et al. Transplantation in HIV + patients. Transplantation, 1990, 49:354–358.
109. Miró JM et al. GESIDA/GESITRA-SEIMC, PNS and ONT consensus document on solid
organ transplant (SOT) in HIV-infected patients in Spain: March 2005. Enfermedades Infecciosas y
Microbiología Clínica, 2005, 23(6):353–362.
110. Ragni MV et al. Survival of human immunodeficiency virus-infected liver transplant recipients. Jour-
nal of Infectious Diseases, 2003, 188(10):1412–1420.
111. Bhopale GM, Nanda RK. Emerging drugs for chronic hepatitis C. Hepatology Research: the Official
Journal of the Japan Society of Hepatology, 2005, 32(3):146–153.
112. De Francesco R, Migliaccio G. Challenges and successes in developing new therapies for hepatitis C.
Nature, 2005, 436(18):953–960.

272

You might also like